1
|
Xu J, Pei Z, Wang Y, Jiang N, Gong Y, Gong F, Ni C, Cheng L. Bioactive microspheres to enhance sonodynamic-embolization-metalloimmune therapy for orthotopic liver cancer. Biomaterials 2025; 317:123063. [PMID: 39753085 DOI: 10.1016/j.biomaterials.2024.123063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 12/07/2024] [Accepted: 12/26/2024] [Indexed: 02/04/2025]
Abstract
The development of novel microspheres for the combination of sonodynamic therapy (SDT) with transarterial embolization (TAE) therapy to amplify their efficacy has received increasing attention. Herein, a novel strategy for encapsulating sonosensitizers (e.g., oxygen-deficient manganese tungstate (MnWOX) nanodots) with gelatin microspheres was proposed. The obtained MnWOX-encapsulated microspheres (abbr. Mn-GMSs) facilitated efficient sonodynamic-embolization-metalloimmune therapy via the immune effects of metal ions on orthotopic liver cancer tumor after transarterial embolization (TAE). Due to the strong cavitation effect caused by the porous structure, Mn-GMSs exhibited a greater reactive oxygen species (ROS) generation rate than the free MnWOX nanodots under US irradiation. Efficient SDT revealed robust cell-killing effects and triggered strong immunogenic cell death (ICD). Moreover, the Mn ions released from the bioactive Mn-GMSs further stimulated the dendritic cells (DCs) maturation and triggered the activation of the cGAS/STING pathway to enhance the immunological effect. Thus, Mn-GMSs achieved significant SDT therapeutic outcomes in H22 tumors in mice, and the combination of the Mn-GMSs triggered SDT with programmed cell death ligand 1 (PD-L1) antibodies could further enhance therapeutic outcomes. The Mn-GMSs exhibited high ROS generation efficacy under US irradiation, significant immune activation, good efficacy in combination with immune checkpoint inhibitor, and great potential for artery embolization-assisted drug delivery, thus enabling effective destruction of liver tumors in rats and rabbits. Therefore, this work provides a strategy for applying SDT in deep tumors and highlights a promising sonodynamic-embolization therapy for combating liver cancers.
Collapse
Affiliation(s)
- Jiachen Xu
- Department of Vascular Surgery and Interventional Radiology, The Forth Affiliated Hospital of Soochow University, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, 215125, China; Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Zifan Pei
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Yuanjie Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Nan Jiang
- Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Yuehan Gong
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Fei Gong
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China.
| | - Caifang Ni
- Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| | - Liang Cheng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
2
|
Ran L, Jiang L, Liu Y, Pang H. Evaluating the efficacy of 8Spheres microsphere embolization combined with iodine‑125 seed implantation in advanced refractory lung cancer: A retrospective study. Oncol Lett 2025; 29:285. [PMID: 40247985 PMCID: PMC12004036 DOI: 10.3892/ol.2025.15031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 03/11/2025] [Indexed: 04/19/2025] Open
Abstract
Patients with advanced non-small cell lung cancer (NSCLC) have seen improvements in care; however, outcomes remain poor for certain individuals despite treatment with radiation, chemotherapy, targeted therapies and immunotherapy. The present study aimed to assess the safety and efficacy of combining 8Spheres microsphere embolization with iodine-125 seed implantation for treating advanced refractory NSCLC. The retrospective analysis included 45 patients with advanced refractory NSCLC. Using the surv_cutpoint function in R, the optimal maximum tumor diameter threshold was determined as 53 mm, dividing patients into two groups: ≤53 mm and >53 mm. The study evaluated the association between treatment regimen, tumor diameter, and progression-free survival (PFS) and overall survival (OS). The findings demonstrated that the experimental group achieved a significantly longer median PFS (12 vs. 10 months; P=0.006) and OS (19 vs. 12 months; P=0.032) compared with the control group. Both the treatment approach and tumor size were identified as independent factors influencing survival. The risk of death was 2.291-fold higher for patients on the control regimen than for those in the experimental group. Similarly, patients with a tumor diameter of >53 mm had a 2.723-fold higher risk of death than those with a tumor diameter of ≤53 mm. Adverse events were mild and resolved in both groups. In summary, the combination of 8Spheres microsphere embolization and iodine-125 seed implantation demonstrate promising clinical outcomes and it may be a viable treatment for advanced refractory NSCLC. Additionally, maximum tumor diameter was strongly associated with patient survival and therefore it may serve as a valuable prognostic indicator to guide treatment decisions.
Collapse
Affiliation(s)
- Linhao Ran
- Department of Nuclear Medicine, Banan People's Hospital, Chongqing Medical University, Chongqing 401320, P.R. China
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Li Jiang
- Department of Ultrasound, Banan People's Hospital, Chongqing Medical University, Chongqing 401320, P.R. China
| | - Ying Liu
- Department of Nuclear Medicine, Banan People's Hospital, Chongqing Medical University, Chongqing 401320, P.R. China
| | - Hua Pang
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
3
|
He M, Xie W, Yuan Z, Chen J, Wang J, Fu Y, Hu Z, Meng Q, Gao W, Hu D, Zhang Y, Pan Y, Zhou Z. Comparing PD-L1 and PD-1 inhibitors plus bevacizumab combined with hepatic arterial interventional therapies in unresetable hepatocellular carcinoma: A single-center, real-world study. Int J Cancer 2025; 156:1972-1985. [PMID: 39834172 DOI: 10.1002/ijc.35341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 12/18/2024] [Accepted: 01/02/2025] [Indexed: 01/22/2025]
Abstract
With the rise of anti-vascular endothelial growth factor antibody and programmed cell death-ligand 1 (PD-L1) regimens, particularly bevacizumab and atezolizumab, as first-line treatments for advanced hepatocellular carcinoma (HCC), there is a need to explore PD-L1 and programmed cell death 1 inhibitors in combination therapies for unresectable HCC (uHCC). Integrating systemic therapies with locoregional approaches is also emerging as a potent strategy. This study compares the outcomes of atezolizumab (PD-L1 inhibitor) and sintilimab (programmed cell death 1 inhibitor) with bevacizumab or its biosimilar, combined with hepatic arterial interventional therapies (HAIT) in uHCC patients. From January 2020 to September 2023, a retrospective analysis was conducted on 138 uHCC patients at Sun Yat-sen University Cancer Center. The cohort included 69 patients treated with atezolizumab with bevacizumab (Bev/Ate) and 69 with bevacizumab biosimilar with sintilimab (Bio/Sin), combined with HAIT. The propensity score matching was also employed to further explore the efficacy and safety. The median progression-free survival (mPFS) was 13.8 months for the Bev/Ate group and 10.0 months for the Bio/Sin group (p = 0.188). The Bev/Ate group showed significantly longer intrahepatic mPFS (HR 0.381; 95% confidence interval 0.176-0.824; p = .018) and higher overall response rates compared with the Bio/Sin group (60.87% vs. 31.88%, p = .001; 69.57% vs. 49.28%, p = .024) based on Response Evaluation Criteria in Solid Tumors v1.1 and modified Response Evaluation Criteria in Solid Tumors criteria. Treatment-related adverse events were similar between groups (p > .050). Combining atezolizumab or sintilimab with bevacizumab or its biosimilar alongside HAIT provided similar overall PFS in uHCC patients. However, the atezolizumab-bevacizumab combination with HAIT showed superior intrahepatic PFS and control rates, warranting further validation.
Collapse
Affiliation(s)
- Minrui He
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, PR China
| | - Wa Xie
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China
- Imaging Diagnostic and Interventional Center, Sun Yat-Sen University Cancer Center, Guangzhou, PR China
| | - Ze Yuan
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China
- Department of Neurosurgery/NeuroOncology, Sun Yat-Sen University Cancer Center, Guangzhou, PR China
| | - Jinbin Chen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, PR China
| | - Juncheng Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, PR China
| | - Yizhen Fu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, PR China
| | - Zili Hu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, PR China
| | - Qi Meng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China
- Department of Clinical Research, Sun Yat-Sen University Cancer Center, Guangzhou, PR China
| | - Wenqing Gao
- Department of Oncology, Tengchong People's Hospital, Baoshan, PR China
| | - Dandan Hu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, PR China
| | - Yaojun Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, PR China
| | - Yangxun Pan
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, PR China
| | - Zhongguo Zhou
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, PR China
| |
Collapse
|
4
|
Qin F, Zheng H, Wu J, Liu Z, Zheng Y, Yang X, Chen J, Deng W, Luo Z, Tan J, Cai W, Jian B, Zeng Y, Qin X, Liao H. APOC1 expressed in macrophages promotes the pulmonary metastasis of colorectal cancer via CCL2/CCL5. Int Immunopharmacol 2025; 154:114611. [PMID: 40194454 DOI: 10.1016/j.intimp.2025.114611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/16/2025] [Accepted: 03/31/2025] [Indexed: 04/09/2025]
Abstract
Metastasis is the main cause of death in colorectal cancer (CRC), and the lungs are common sites of metastasis. However, there is little effective target to intervene colorectal cancer pulmonary metastasis (CCPM), especially on its unique immune microenvironment. In this study, sixteen genes were identified as core CCPM-related differentially expressed genes (DEGs) between CRC and CCPM. Three genes including Apolipoprotein C1 (APOC1) were associated with prognosis, stage and metastasis of CRC. In immunohistochemistry, APOC1 was mainly expressed in macrophages, and expressed more in CCPM than CRC. Patients with synchronous CCPM, higher stage, poorer OS and CCPM-free interval tended to have higher expression. In experiments in vitro, knockdown of APOC1 in macrophages reduced the migration, invasion, and epithelial-mesenchymal transition of CRC cells. Knockdown of APOC1 in macrophages significantly decreased secretion of chemokines like CCL2 and CCL5. The pro-metastatic effect of macrophages expressing APOC1 was partially blocked by the antibodies of CCL2 and CCL5. Activation of STAT3 was a key process in APOC1's regulation of CCL2 and CCL5. In experiments in vivo, knockdown of APOC1 in macrophages reduced pulmonary metastasis. To conclude, APOC1 is one of core CCPM-related DEGs and associated with the metastasis and survival of CRC. Macrophages expressing APOC1 promote the CCPM by APOC1-STAT3-CCL2/CCL5 axis. APOC1 and macrophages expressing APOC1 play vital roles and may be potential therapeutic targets in CCPM.
Collapse
Affiliation(s)
- Fei Qin
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Haosheng Zheng
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Jiayan Wu
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Zui Liu
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Yuzhen Zheng
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Xingping Yang
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Junguo Chen
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Weihao Deng
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Ziyin Luo
- Department of Otorhinolaryngology Head and Neck Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Jian Tan
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Weijie Cai
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Bozhu Jian
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Yushuai Zeng
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Xianyu Qin
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China.
| | - Hongying Liao
- Department of Thoracic Surgery, Thoracic Cancer Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China.
| |
Collapse
|
5
|
Hohenberger P, Rathmann N, Büsing K, Menge F, Jakob J, Pink D, Wardelmann E, Schoenberg SO, Diehl SJ. Selective internal radiation with Y-90 resin microspheres (SIRT) for liver metastases of gastro-intestinal stromal tumors (GIST) resistant to tyrosine kinase inhibitor (TKI) therapy. Br J Cancer 2025; 132:716-724. [PMID: 40044980 PMCID: PMC11997030 DOI: 10.1038/s41416-025-02952-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 12/20/2024] [Accepted: 01/31/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Hepatic metastases of GIST might be the dominant site of progression and resistant to available tyrosine kinase inhibitors (TKIs). Selective internal radiation therapy (SIRT) offers treatment by intratumoral radiation up to 200 Gy. We analyzed the hepatic progression-free survival (H-PFS) in a consecutive patient cohort. METHODS Twenty-six patients (median age 57.6 years) with biopsy proven liver metastases of GIST were treated by SIRT. All had RECIST documented tumor progression, and 24/26 patients had up to four lines of pretreatment. Mutational status was 'quadruple wildtype' (q-wt, n = 5), KIT exon 11/9/13 in n = 15/4/1 cases and PDGFRα (n = 1). Median follow-up of this retrospective analysis of a prospectively kept database is 33.6 months. RESULTS Median H-PFS was 16 months (range, 4-54+ months, 95% CI 6.5-25.4 months) and OS after SIRT was 28 months (95% CI 17.2-28.7 months). Best H-PFS was observed in patients with 'q-wt' at 25 months (range, 6+-54 months, 95% CI 16.2-33.8 months). The worst outcome was for KIT exon 11 mutations plus secondary mutations with 7 months (range, 4-33 months, 95% CI, 4.2-9.8 months). CONCLUSIONS 90Y-SIRT is a potent treatment for patients with liver metastases of GIST resistant to TKI therapy. In patients with 'q-wt' GIST, SIRT is an option for first-line use.
Collapse
Affiliation(s)
- Peter Hohenberger
- Division of Surgical Oncology and Thoracic Surgery, University Medical Center Mannheim, Medical Faculty Mannheim - Heidelberg University, Heidelberg, Germany.
| | - Nils Rathmann
- Institute of Clinical Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim - Heidelberg University, Heidelberg, Germany
| | - Karen Büsing
- Institute of Clinical Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim - Heidelberg University, Heidelberg, Germany
| | - Franka Menge
- Division of Surgical Oncology and Thoracic Surgery, University Medical Center Mannheim, Medical Faculty Mannheim - Heidelberg University, Heidelberg, Germany
| | - Jens Jakob
- Department of Surgery Mannheim University Medical Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Daniel Pink
- Sarkomzentrum Berlin-Brandenburg, HELIOS Klinikum Berlin-Buch, Berlin, Germany
| | - Eva Wardelmann
- Sarkomzentrum Berlin-Brandenburg, HELIOS Klinikum Berlin-Buch, Berlin, Germany
- Gerhard Domagk Institute of Pathology, University Hospital Muenster, Muenster, Germany
| | - Stefan O Schoenberg
- Institute of Clinical Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim - Heidelberg University, Heidelberg, Germany
| | - Steffen J Diehl
- Institute of Clinical Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim - Heidelberg University, Heidelberg, Germany
| |
Collapse
|
6
|
Bucher HC, Chammartin F. Strengthening health technology assessment for cancer treatments in Europe by integrating causal inference and target trial emulation. THE LANCET REGIONAL HEALTH. EUROPE 2025; 52:101294. [PMID: 40255411 PMCID: PMC12008668 DOI: 10.1016/j.lanepe.2025.101294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 03/16/2025] [Accepted: 03/21/2025] [Indexed: 04/22/2025]
Abstract
Health Technology Assessment (HTA) for reimbursement of all new cancer drugs in the European Union (EU) will be evaluated for all member states by a central European HTA starting in 2025. EU HTA guidelines for applicants under these new regulations put the focus on meta-analysis of aggregated randomized trial data and are in contrast with the growing number of cancer drug approvals by drug regulators that are based on single arm studies and the needs in the rapidly evolving field of oncological drug development. We advocate to broaden the methodological approaches for HTA by including observational data based causal inference methodology and target trial emulation into the assessments of comparative effectiveness. Causal inference estimates causal estimands, effect measures that reflect a population level effect in terms of contrasts of counterfactual outcomes in the same patients and are directly measured in the target population by modeling of hypothetical intervention. Target trial emulation allows with the use of causal inference to estimate causal effects by mimicking pragmatic trials that evolve apart from randomization like a trial. We illustrate the potential of causal inference for HTA and provide an introduction into causal inference methodology for health scientists involved in HTA.
Collapse
Affiliation(s)
- Heiner C. Bucher
- Division of Clinical Epidemiology, Department of Clinical Research, University Hospital and University of Basel, Basel, Switzerland
| | - Frédérique Chammartin
- Division of Clinical Epidemiology, Department of Clinical Research, University Hospital and University of Basel, Basel, Switzerland
| |
Collapse
|
7
|
Zhou S, Song C, Liu P, Ju S, Wang YC. A nationwide investigation on imaging follow-up after Locoregional therapy for hepatocellular carcinoma in China: Current practices and challenges. Eur J Radiol 2025; 186:112057. [PMID: 40132470 DOI: 10.1016/j.ejrad.2025.112057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 02/22/2025] [Accepted: 03/17/2025] [Indexed: 03/27/2025]
Abstract
PURPOSE To investigate the perspectives of interventional radiologists in China on imaging follow-up protocols and adherence to treatment response criteria for hepatocellular carcinoma (HCC) following locoregional therapies (LRT), with a particular focus on identifying gaps and proposing strategies to bridge the discrepancy between clinical guidelines and real-world practice. MATERIALS AND METHODS The web-based survey was conducted among members of Chinese Society of Radiology and Zhongda Radiology Alliance between 1st October 2024 to 30th November 2024, via WPS Office survey tool. The frequencies and percentages of responses were summarized as n (%), and a chi-squared test was employed to compare the responses among diagnostic radiologists, interventional radiologists, and interventional physicians. RESULTS A total of 604 respondents from 325 hospitals in 31 provinces were analyzed. Of the respondents, 72.7 % (439/604) believed that the same imaging modality should be used for follow-up after conventional transarterial chemoembolization (cTACE) and other LRT for HCC. Among these respondents, contrast-enhanced computed tomography (CE-CT) (57.2 %, 251/439) was the most preferred imaging modality for initial follow-up, and hepatobiliary contrast-enhanced MRI (63.1 %, 277/439) for subsequent follow-up examinations. For respondents (27.3 %, 165/604) who believed that follow-up strategies should be tailored to the type of LRT, CE-CT was most commonly recommended for post-cTACE HCC, both for initial (64.2 %, 106/165) and subsequent (60.6 %, 100/165) follow-up. For HCC treated with other LRT, the majority of respondents preferred extracellular contrast-enhanced MRI for initial follow-up (55.8 %, 92/165) and hepatobiliary contrast-enhanced MRI for subsequent follow-up (61.2 %, 101/165). The most recommended time frame for initial follow-up was "within 1-2 months" among all respondents. However, significant differences in the recommended time frame were observed among diagnostic radiologists, interventional radiologists, and interventional physicians (P < 0.001). Notably, more than 95 % of the respondents who selected 'unclear' were diagnostic radiologists. The most recommended monitoring frequency was every 3-4 months (46.0 %, 278/604) for viable lesions and every 5-6 months (32.9 %, 199/604) for nonviable lesions. Regarding adherence to treatment response criteria, mRECIST (32.0 %, 193/604) and LR-TRA (v2017/v2024) (24.2 %, 146/604) were commonly adopted in clinical practice. Nevertheless, a significant proportion of respondents (25.2 %, 152/604) indicated that none of these criteria were applied in their clinical practice. CONCLUSION CE-CT performed within 1-2 months was the most preferred modality and time frame for initial follow-up. Significant variability remains in follow-up frequency and treatment response criteria for post-LRT HCC, highlighting the need for further standardization of imaging follow-up protocols and structured treatment response assessment to optimize post-LRT management in clinical practice.
Collapse
Affiliation(s)
- Shuwei Zhou
- Department of Radiology, Zhongda Hospital, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, School of Medicine, Southeast University, Nanjing 210009, China.
| | - Chenxin Song
- Department of Radiology, Zhongda Hospital, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, School of Medicine, Southeast University, Nanjing 210009, China.
| | - Pei Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Southeast University, Nanjing 210009, China.
| | - Shenghong Ju
- Department of Radiology, Zhongda Hospital, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, School of Medicine, Southeast University, Nanjing 210009, China.
| | - Yuan-Cheng Wang
- Department of Radiology, Zhongda Hospital, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, School of Medicine, Southeast University, Nanjing 210009, China.
| |
Collapse
|
8
|
Lu X, Friedrich LJ, Efferth T. Natural products targeting tumour angiogenesis. Br J Pharmacol 2025; 182:2094-2136. [PMID: 37680009 DOI: 10.1111/bph.16232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/15/2023] [Accepted: 08/28/2023] [Indexed: 09/09/2023] Open
Abstract
Tumour angiogenesis is the formation of new blood vessels to support the growth of a tumour. This process is critical for tumour progression and metastasis, making it an attractive approach to cancer therapy. Natural products derived from plants, animals or microorganisms exert anti-angiogenic properties and can be used to inhibit tumour growth and progression. In this review, we comprehensively report on the current status of natural products against tumour angiogenesis from four perspectives until March 2023: (1) the role of pro-angiogenic factors and antiangiogenic factors in tumour angiogenesis; (2) the development of anti-tumour angiogenesis therapy (monoclonal antibodies, VEGFR-targeted small molecules and fusion proteins); (3) the summary of anti-angiogenic natural agents, including polyphenols, polysaccharides, alkaloids, terpenoids, saponins and their mechanisms of action, and (4) the future perspectives of anti-angiogenic natural products (bioavailability improvement, testing of dosage and side effects, combination use and discovery of unique natural-based compounds). Our review aims to better understand the potential of natural products for drug development in inhibiting tumour angiogenesis and further aid the effective transition of these outcomes into clinical trials. LINKED ARTICLES: This article is part of a themed issue Natural Products and Cancer: From Drug Discovery to Prevention and Therapy. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v182.10/issuetoc.
Collapse
Affiliation(s)
- Xiaohua Lu
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Lara Johanna Friedrich
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
9
|
Zhang L, Wang D, Zhang LZ, Yang WH, Yu C, Qin J, Feng LZ, Liu Z, Teng GJ. Pickering emulsion with tumor vascular destruction and microenvironment modulation for transarterial embolization therapy. Biomaterials 2025; 316:123018. [PMID: 39709852 DOI: 10.1016/j.biomaterials.2024.123018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 12/02/2024] [Accepted: 12/13/2024] [Indexed: 12/24/2024]
Abstract
In the clinic, Lipiodol chemotherapeutic emulsions remain a main choice for patients diagnosed with hepatocellular carcinoma (HCC) via the mini-invasive transarterial chemoembolization (TACE) therapy. However, the poor stability of conventional Lipiodol chemotherapeutic emulsions would result in the fast drug diffusion and incomplete embolization, inducing systemic toxicity and impairing the efficacy of TACE therapy. Therefore, it is of great importance to construct alternative formulations based on commercial Lipiodol to achieve the improved efficacy and safety of HCC treatment. Herein, calcium phosphate (CaP) nanoparticles-stabilized Lipiodol Pickering emulsion (CaP-LPE) with improved stability and pH-responsiveness is prepared and utilized for the encapsulation of combretastatin A4-phosphate (CA4P), a clinically approved vascular disrupting agent. The obtained CA4P-loaded CaP-LPE (CCaP-LPE) was shown to be enhanced stability compared to conventional Lipiodol emulsion and pH-responsive release of the encapsulated drugs. On one hand, the released CA4P could disrupt tumor vascular and cut off the blood supplying of tumor cells, thus starving cancer cells. Moreover, it was revealed that CCaP-LPE could reverse immunosuppressive tumor microenvironment (TME) by neutralizing tumor acidity, leading to the increased infiltration of CD8+ T cells and the decreased percentages of immunosuppressive cells. As the result, such CCaP-LPE could effectively shrink orthotopic N1S1 HCC tumors in rats by eliciting a potent antitumor immune response. Therefore, this study highlights a simple strategy to construct a novel LPE with the potencies of tumor vascular disruption and TME modulation, holding a great promise for TAE therapy of HCC.
Collapse
Affiliation(s)
- Lei Zhang
- Center of Interventional Radiology and Vascular Surgery, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Department of Radiology, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China; National Innovation Platform for Integration of Medical Engineering Education (NMEE) (Southeast University), Nanjing, 210009, China; Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing, 210009, China; State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, 210009, China
| | - Duo Wang
- Center of Interventional Radiology and Vascular Surgery, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Department of Radiology, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China; National Innovation Platform for Integration of Medical Engineering Education (NMEE) (Southeast University), Nanjing, 210009, China; Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing, 210009, China; State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, 210009, China
| | - Lin-Zhu Zhang
- Center of Interventional Radiology and Vascular Surgery, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Department of Radiology, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China; National Innovation Platform for Integration of Medical Engineering Education (NMEE) (Southeast University), Nanjing, 210009, China; Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing, 210009, China; State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, 210009, China
| | - Wei-Hao Yang
- Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Chao Yu
- Center of Interventional Radiology and Vascular Surgery, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Department of Radiology, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China; National Innovation Platform for Integration of Medical Engineering Education (NMEE) (Southeast University), Nanjing, 210009, China; Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing, 210009, China; State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, 210009, China
| | - Juan Qin
- Center of Interventional Radiology and Vascular Surgery, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Department of Radiology, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China; National Innovation Platform for Integration of Medical Engineering Education (NMEE) (Southeast University), Nanjing, 210009, China; Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing, 210009, China; State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, 210009, China
| | - Liang-Zhu Feng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China.
| | - Gao-Jun Teng
- Center of Interventional Radiology and Vascular Surgery, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Department of Radiology, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China; National Innovation Platform for Integration of Medical Engineering Education (NMEE) (Southeast University), Nanjing, 210009, China; Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing, 210009, China; State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, 210009, China.
| |
Collapse
|
10
|
van Dorp M, Gonzalez M, Ojanguren A, Brunelli A. Transcontinental Differences in Management of Pulmonary Metastatic Disease: Europe. Thorac Surg Clin 2025; 35:233-247. [PMID: 40246413 DOI: 10.1016/j.thorsurg.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
Several international registries, including 4 from Europe-Italy, Spain, the Netherlands, and the European Society of Thoracic Surgeons-are dedicated to addressing deficiencies in pulmonary metastasectomy research. The randomized PulMiCC and SABR-COMET trials provide contradictory outcomes and new randomized trials have been initiated. Europe's metastasectomy approach transitioned from open thoracotomy to video-assisted thoracoscopic surgery, with 72% of surgeons favoring minimally invasive methods by 2023. European Society for Medical Oncology guidelines recommend surgery for completely resectable lesions and propose ablative approaches as supplementary or alternative treatments for inoperable cases due to frailty or unfavorable anatomy.
Collapse
Affiliation(s)
- Martijn van Dorp
- Department of Cardiothoracic Surgery, Amsterdam University Medical Center - Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Michel Gonzalez
- Division of Thoracic Surgery, Lausanne University Hospital, Lausanne, Switzerland
| | - Amaia Ojanguren
- Department of Thoracic Surgery, Bellvitge University Hospital, Barcelona, Spain
| | - Alessandro Brunelli
- Department of Thoracic Surgery, St James's University Hospital, Beckett Street, Leeds LS9 7TF, UK.
| |
Collapse
|
11
|
Li Q, Chang X, Gu J, Yang Y, Ouyang J, Zhou Y, Zhao H, Zhou J. Adjuvant Transarterial Chemoembolization in Resected Macrotrabecular-massive Hepatocellular Carcinoma (ATAC-MACRO): A Multicenter Real-world Retrospective Study. Acad Radiol 2025; 32:2585-2595. [PMID: 39848885 DOI: 10.1016/j.acra.2024.12.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 12/08/2024] [Accepted: 12/22/2024] [Indexed: 01/25/2025]
Abstract
RATIONALE AND OBJECTIVES The purpose of this study was to demonstrate the impact of postoperative adjuvant transarterial chemoembolization (TACE) on the prognosis of patients with macrotrabecular-massive hepatocellular carcinoma (MTM-HCC). MATERIALS AND METHODS This retrospective study used the clinical records of patients with resected MTM-HCC with/without adjuvant TACE at three centers between January 2015 and December 2022. The primary end point was recurrence free survival (RFS). The secondary end points were overall survival (OS) and safety. RESULTS A total of 559 eligible patients were classified into the adjuvant TACE group and the observation group. After propensity score matching analysis, both RFS (HR 0.62 [95% CI, 0.48 to 0.80]; P < 0.001) and OS (HR 0.59 [95% CI, 0.42 to 0.84]; P = 0.013) in the adjuvant TACE group were significantly better than the observation group. By Cox regression models, mALBI grade, types of hepatectomy, number, satellite lesion, without adjuvant TACE were identified as independent risk factors for RFS, and mALBI grade, number, maximum tumor size, satellite lesion, microvascular invasion, high AFP level, without adjuvant TACE were identified as independent risk factors for OS. The incidence of surgery-related adverse events (AEs) had no significant difference between the two groups (P = 0.609). The majority of AEs associated with adjuvant TACE were grade I (84.4%), and no treatment-related deaths occurred in either group. CONCLUSIONS Adjuvant TACE significantly improved the RFS and OS of patients with resected MTM-HCC with acceptable toxicity.
Collapse
Affiliation(s)
- Qingjun Li
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China (Q.L., J.O., Y.Z., J.Z.)
| | - Xu Chang
- Department of Interventional Therapy II, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China (X.C.)
| | - Jiaye Gu
- School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China (J.G.)
| | - Yi Yang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (Y.Y., H.Z.)
| | - Jingzhong Ouyang
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China (Q.L., J.O., Y.Z., J.Z.)
| | - Yanzhao Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China (Q.L., J.O., Y.Z., J.Z.)
| | - Hong Zhao
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (Y.Y., H.Z.)
| | - Jinxue Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China (Q.L., J.O., Y.Z., J.Z.).
| |
Collapse
|
12
|
Ye X, Fang X, Li F, Jin D. Targeting TIME in Advanced Hepatocellular Carcinoma: Mechanisms of Drug Resistance and Treatment Strategies. Crit Rev Oncol Hematol 2025:104735. [PMID: 40250780 DOI: 10.1016/j.critrevonc.2025.104735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/04/2025] [Accepted: 04/12/2025] [Indexed: 04/20/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is the predominant form of primary liver cancer. While early-stage HCC can be effectively managed with surgical resection and other interventions, treatment options for advanced HCC are limited. Current systemic treatments for advanced HCC include VEGF-targeted tyrosine kinase inhibitors (Sorafenib, Lenvatinib), and the combination therapy of anti PD-1/PD-L1 and anti VEGF (Atezolizumab plus Bevacizumab, Camrelizumab plus Rivoceranib). However, the lack of response to these drugs and the emergence of acquired drug resistance significantly impairs their efficacy. Numerous studies have demonstrated that the tumor immune microenvironment (TIME) plays a crucial role in modulating the response to these therapies. Various immune cells and their secreted factors within the TIME play a pivotal role in the emergence of secondary drug resistance in HCC. This article reviews the mechanism of TIME promoting drug resistance, discusses the influence of current systemic HCC treatment drugs on TIME, and evaluates how these TIME changes affect the efficacy of treatment. A deeper understanding of the interaction between TIME and systemic treatment drugs may be beneficial to enhance the treatment effect, mitigate drug resistance of advanced HCC, and ultimately improve the prognosis of patients.
Collapse
Affiliation(s)
- Xinyi Ye
- Department of Immunology and Pathogenic Biology, Yanbian University Medical College, Yanji, 13002, China.
| | - Xizhu Fang
- Department of Immunology and Pathogenic Biology, Yanbian University Medical College, Yanji, 13002, China.
| | - Fangfang Li
- Department of Immunology and Pathogenic Biology, Yanbian University Medical College, Yanji, 13002, China.
| | - Dan Jin
- Department of Immunology and Pathogenic Biology, Yanbian University Medical College, Yanji, 13002, China.
| |
Collapse
|
13
|
Miao YR, Yang XJ. Hepatocellular carcinoma resistance to tyrosine kinase inhibitors: Current status and perspectives. World J Gastrointest Oncol 2025; 17:101528. [PMID: 40235904 PMCID: PMC11995346 DOI: 10.4251/wjgo.v17.i4.101528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 12/31/2024] [Accepted: 01/08/2025] [Indexed: 03/25/2025] Open
Abstract
The study conducted by Wang et al, focuses on the role of Rho GTPase activating protein 12 (ARHGAP12), in hepatocellular carcinoma (HCC). This research reveals that ARHGAP12 expression, markedly elevated in malignant cells of HCC, correlates strongly with adverse outcomes for patients. Furthermore, the study illustrates that ARHGAP12 enhances the ability of HCC cells to invade and contributes to their resistance to tyrosine kinase inhibitors (TKIs) through modulation of the focal adhesion pathway. To comprehensively investigate the relationship between ARHGAP12 and TKI resistance, this study integrates single-cell and bulk RNA sequencing methodologies along with data from tumor immune single-cell hub 2, Gene Expression Omnibus, The Cancer Genome Atlas, CellMiner, Genomics of Drug Sensitivity in Cancer 2, as well as immunohistochemical staining and proteomic analyses. Statistical analyses, including the Wilcoxon rank-sum test and receiver operating characteristic curve analysis, were employed to evaluate the correlation between ARHGAP12 expression levels and clinical parameters, as well as drug sensitivity. It is evident that a more profound exploration of the molecular dynamics of HCC, especially those related to resistance against TKIs, is essential.
Collapse
Affiliation(s)
- Yu-Run Miao
- The First Clinical Medical School, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China
- Second Ward of General Surgery, Gansu Province People Hospital, Lanzhou 730000, Gansu Province, China
| | - Xiao-Jun Yang
- Second Ward of General Surgery, Gansu Province People Hospital, Lanzhou 730000, Gansu Province, China
| |
Collapse
|
14
|
Fu Y, Wu J, Wu J, Li Y, Zeng Z, Liu D, Li H, Ou X, Lin Z, Wei S, Song H, Yan M. Combination Therapy of Transarterial Chemoembolization, Lenvatinib, and PD-1 Inhibitors Achieves Significant Tumor Response in Hepatocellular Carcinoma with Bile Duct Tumor Thrombus: A Case Report. Cancer Manag Res 2025; 17:793-799. [PMID: 40256770 PMCID: PMC12007008 DOI: 10.2147/cmar.s511319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 04/04/2025] [Indexed: 04/22/2025] Open
Abstract
Combination therapy plays a critical role in optimizing surgical outcomes for patients with locally advanced hepatocellular carcinoma (HCC) complicated by bile duct tumor thrombus (BDTT). Current neoadjuvant strategies integrate local and systemic modalities to reduce tumor burden and recurrence rate. However, the combination of transarterial chemoembolization (TACE), lenvatinib, and PD-1 inhibitors (triple therapy) as a neoadjuvant regimen for HCC with BDTT has not been previously reported. Here, we present the case of a 61-year-old man with HBV-associated HCC and BDTT, initially deemed high-risk for direct resection due to tumor size (7 cm) and biliary involvement. The patient underwent one session of TACE followed by two months of lenvatinib (12 mg/day) and sintilimab (200 mg every 3 weeks). Post-treatment contrast-enhanced MRI revealed complete resolution of BDTT and partial response of the primary tumor. Subsequent right hemihepatectomy confirmed extensive tumor necrosis (>90%) with negative margins. At 15-month follow-up, surveillance imaging showed no recurrence. The patient experienced only grade 1 hypertension, managed without treatment interruption. This case highlights the potential of triple therapy as a neoadjuvant approach to downstage advanced HCC with BDTT, enabling curative resection while maintaining a manageable safety profile. Further studies are warranted to validate its efficacy in larger cohorts and define optimal treatment protocols.
Collapse
Affiliation(s)
- Yangkai Fu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian Province, People’s Republic of China
| | - Junyi Wu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian Province, People’s Republic of China
- Department of Hepatobiliary Pancreatic Surgery, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian Province, People’s Republic of China
| | - Jiayi Wu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian Province, People’s Republic of China
- Department of Hepatobiliary Pancreatic Surgery, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian Province, People’s Republic of China
| | - Yinan Li
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian Province, People’s Republic of China
| | - Zhenxin Zeng
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian Province, People’s Republic of China
| | - Deyi Liu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian Province, People’s Republic of China
| | - Han Li
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian Province, People’s Republic of China
| | - Xiangye Ou
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian Province, People’s Republic of China
| | - Zhongtai Lin
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian Province, People’s Republic of China
| | - Shaoming Wei
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian Province, People’s Republic of China
| | - Huachun Song
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian Province, People’s Republic of China
| | - Maolin Yan
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian Province, People’s Republic of China
- Department of Hepatobiliary Pancreatic Surgery, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian Province, People’s Republic of China
| |
Collapse
|
15
|
Peng Y, Liu H, Liang X, Cao L, Teng M, Chen H, Li Z, Peng X, Mao J, Cheng H, Liu G. Self-assembling chemodrug fiber-hydrogel for transarterial chemoembolization and radiotherapy-enhanced antitumor immunity. J Control Release 2025; 380:1-16. [PMID: 39892652 DOI: 10.1016/j.jconrel.2025.01.088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/25/2025] [Accepted: 01/28/2025] [Indexed: 02/04/2025]
Abstract
Hydrogel, as a promising embolic material for hepatocellular carcinoma (HCC), may fully embolize both major vessels and peripheral microvessels. A self-assembling hydrogel composed of chemotherapeutic drugs offers significant clinical benefits without carrier introduction. Herein, we developed a sustained drug-releasing complex hydrogel (RKT@gel), which was fabricated by the self-assembly of raltitrexed chemotherapeutic drugs (R@gel), along with the incorporation of kaempferol and tantalum nanoparticles (Ta NPs). Kaempferol enhances the mechanical strength of R@gel and inhibits hypoxia-induced angiogenesis post-embolization, improving embolization effectiveness. In addition to enabling X-ray-guided transarterial chemoembolization (TACE), Ta NPs enhance radiation sensitivity. These synergistic effects of RKT@gel not only significantly induce immunogenic cell death, thereby enhancing the activation of dendritic cells, but also activate major histocompatibility complex class I (MHC-I)-mediated antitumor immune recognition and cytotoxicity. In vivo, RKT@gel achieves enhanced tumor deposition and sustained drug release, effectively suppressing tumor progression. Additionally, when combined with radiotherapy, RKT@gel achieves efficient antitumor immunoactivation. Overall, this versatile composite hydrogel not only achieves effective embolization therapy but also substantially triggers antitumor immune responses with good biocompatibility. This multifunctional design provides a TACE-based multidisciplinary strategy for HCC.
Collapse
Affiliation(s)
- Yisheng Peng
- State Key Laboratory of Vaccine for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, Fujian Engineering Research Center of Molecular Theranostic Technology, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Hui Liu
- State Key Laboratory of Vaccine for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, Fujian Engineering Research Center of Molecular Theranostic Technology, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Xiaoliu Liang
- College of Pharmacy, Guangxi Medical University, Nanning 530021, China
| | - Lei Cao
- Department of Pathology, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen 361102, China
| | - Minglei Teng
- State Key Laboratory of Vaccine for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, Fujian Engineering Research Center of Molecular Theranostic Technology, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Hu Chen
- State Key Laboratory of Vaccine for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, Fujian Engineering Research Center of Molecular Theranostic Technology, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Zhenjie Li
- State Key Laboratory of Vaccine for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, Fujian Engineering Research Center of Molecular Theranostic Technology, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Xuqi Peng
- State Key Laboratory of Vaccine for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, Fujian Engineering Research Center of Molecular Theranostic Technology, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Jingsong Mao
- State Key Laboratory of Vaccine for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, Fujian Engineering Research Center of Molecular Theranostic Technology, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Hongwei Cheng
- State Key Laboratory of Vaccine for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, Fujian Engineering Research Center of Molecular Theranostic Technology, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China; Zhuhai UM Science & Technology Research Institute, University of Macau, Macau 999078, China.
| | - Gang Liu
- State Key Laboratory of Vaccine for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, Fujian Engineering Research Center of Molecular Theranostic Technology, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
16
|
Zhan J, Huang H, Zhan T, Liu X, Cheng Q. Rare Gingival Metastasis Occurring After Conversion Therapy Followed by Resection of Initially Unresectable Hepatocellular Carcinoma: A Case Report. J Hepatocell Carcinoma 2025; 12:705-713. [PMID: 40226819 PMCID: PMC11994111 DOI: 10.2147/jhc.s514983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Accepted: 03/31/2025] [Indexed: 04/15/2025] Open
Abstract
Gingival metastases from hepatocellular carcinoma (HCC) are exceedingly rare and highly prone to be misdiagnosed without biopsy. Here, we report an initially unresectable HCC patient who received effective conversion therapy but discovered gingival metastasis within one-month post-hepatectomy. A 53-year-old male with a huge liver tumor diagnosed as unresectable HCC received conversion therapy of hepatic arterial infusion chemotherapy (HAIC) combined with lenvatinib and tislelizumab. During the conversion therapy, he experienced sore gingiva which was regarded as a side effect of lenvatinib. Considering the significant shrinkage of tumor after 10-month treatment, salvage resection was conducted with negative margin and no postoperative complications. Gingival oligometastases were identified and resected half month after surgery. Throughout the 1-year follow-up period, the patient remained alive; however, there was a recurrence of the gingival metastasis at the same site six months postoperatively. Hence, clinicians should regard gingival swelling and pain not merely as potential adverse events of conversion therapy but also as potential indicators of gingival metastasis.
Collapse
Affiliation(s)
- Juncheng Zhan
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, People’s Republic of China
- Department of Clinical Medicine, Bengbu Medical University, Bengbu, Anhui Province, 233004, People’s Republic of China
| | - Hongwei Huang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, People’s Republic of China
| | - Tianao Zhan
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, People’s Republic of China
| | - Xinkang Liu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, People’s Republic of China
| | - Qi Cheng
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, People’s Republic of China
| |
Collapse
|
17
|
Tian H, Wan C. Comparative efficacy and safety of transarterial chemoembolization combined with tyrosine kinase inhibitors and immune checkpoint inhibitors versus tyrosine kinase inhibitors and immune checkpoint inhibitors alone in advanced hepatocellular carcinoma: a systematic review and meta-analysis. World J Surg Oncol 2025; 23:126. [PMID: 40197348 PMCID: PMC11974228 DOI: 10.1186/s12957-025-03788-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 03/29/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a leading cause of cancer-related mortality worldwide, and advanced-stage disease presents significant therapeutic challenges. Combining transarterial chemoembolization (TACE) with tyrosine kinase inhibitors (TKIs) and immune checkpoint inhibitors (ICIs) has emerged as a promising strategy to enhance treatment efficacy. This meta-analysis evaluates efficacy and safety of TACE + TKIs + ICIs compared to TKIs + ICIs alone in patients with HCC. METHODS A systematic search was conducted across "PubMed", "Web of Science", "Cochrane Library", "Scopus", "Google Scholar", and "Embase" to screen studies up to November 2024. Studies comparing TACE + TKIs + ICIs with TKIs + ICIs alone in advanced HCC were included. Outcomes of interest included objective response rate (ORR), disease control rate (DCR), overall survival (OS), progression-free survival (PFS), and adverse events. Results were reported as relative risk (RR) or hazard ratios (HR) with 95% confidence intervals (CI). Funnel plots was used to assess publication bias. RESULTS Ten studies comprising 1999 patients were included. The combination of TACE + TKIs + ICIs marked improved ORR (RR = 1.81, 95%CI:1.57-2.09, P < 0.00001) and DCR (RR = 1.32, 95%CI: 1.19-1.46, P < 0.00001) comparing with TKIs + ICIs alone. OS and PFS were also significantly prolonged in combination group, with HR of 0.55 (95%CI:0.48-0.63, P < 0.00001) and 0.73 (95%CI:0.65-0.82, P < 0.00001), respectively. Adverse events such as pain (RR = 3.94, 95%CI:2.40-6.47, P < 0.001) and nausea/vomiting (RR = 2.28, 95% CI:1.56-3.33, P < 0.001) were more frequent in the TACE + TKIs + ICIs group, though rates of hypertension, diarrhea, and rash were similar between groups. Funnel plots indicated minimal publication bias for primary outcomes. CONCLUSIONS The combination of TACE, TKIs, and ICIs significantly improves ORR, DCR, OS, and PFS compared to TKIs and ICIs alone, demonstrating superior efficacy with an acceptable safety profile. These findings provide evidence for the integration of TACE with systemic therapies in the management of HCC.
Collapse
Affiliation(s)
- Hengyu Tian
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, No, China
| | - Chidan Wan
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, No, China.
| |
Collapse
|
18
|
Wang G, Pan Y, Zheng L, Zhang X, Liu H, Xu Y, Zhang W, Luan X, Liu X, Xu X, Wu S, Ma G, Kan Y, Zhang J, Wang R, Yang J. Monitoring Sorafenib Resistance and Efficacy in Hepatocellular Carcinoma Using [ 18F]Alfatide II and [ 18F]Fluorodeoxyglucose Positron Emission Tomography. Mol Pharm 2025; 22:2088-2097. [PMID: 39988972 DOI: 10.1021/acs.molpharmaceut.4c01218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Integrin αvβ3 expression is associated with sorafenib resistance in hepatocellular carcinoma (HCC). Therefore, monitoring its expression in HCC may serve as a valuable indicator of the efficacy of sorafenib treatment. In this study, longitudinal positron emission tomography (PET) was performed to assess [18F]Alfatide II and [18F]fluorodeoxyglucose ([18F]FDG) as suitable probes for evaluating the treatment efficacy of sorafenib in a Huh-7 human (HCC) xenograft model. Huh-7 tumor cells were used to establish both normal and sorafenib-resistant cell lines, and xenograft models were developed. The mice were categorized into four groups based on the cell type and treatment: normal nontreatment, normal treatment, sorafenib-resistant nontreatment, and sorafenib-resistant treatment. Huh-7 tumor mice received intragastric injections of sorafenib (30 mg/kg/day) or vehicle for 15 consecutive days. Tumor size and weight were assessed throughout the study. Longitudinal microPET/computed tomography (CT) scans with [18F]Alfatide II and [18F]FDG were acquired to quantitatively measure angiogenesis on days -2, 3, 7, and 14 and metabolism on days -1, 4, 8, and 15 following therapy initiation. The tumor uptake (ID%/gmean) of each probe was calculated. No significant difference in [18F]FDG uptake was observed between the normal and sorafenib-resistant groups (P = 0.452); however, [18F]Alfatide II uptake differed significantly between the two groups (P < 0.001). Sorafenib successfully inhibited normal Huh-7 tumor growth, inducing significant differences in tumor size 9 days after sorafenib treatment (P < 0.05). The uptake of [18F]Alfatide II in the tumor lesions changed significantly on day 14 (P = 0.001). However, no change was observed in the uptake of [18F]FDG (P > 0.05). The PET imaging data of [18F]Alfatide II and [18F]FDG were validated through ex vivo immunohistochemistry analysis targeting integrin αvβ3, VEGF, and GULT-1. [18F]Alfatide II PET was more effective in monitoring sorafenib resistance and therapeutic efficacy in the Huh-7 human HCC xenograft model than [18F]FDG.
Collapse
Affiliation(s)
- Guanyun Wang
- Nuclear Medicine Department, Beijing Friendship Hospital, Capital Medical University, 95 Yong'an Road, Xicheng District, Beijing 100050, China
| | - Yue Pan
- Department of Nuclear Medicine, The First Medical Center, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing 100853, China
- Graduate School, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Lingling Zheng
- Nuclear Medicine Department, Beijing Friendship Hospital, Capital Medical University, 95 Yong'an Road, Xicheng District, Beijing 100050, China
| | - Xiaojun Zhang
- Department of Nuclear Medicine, The First Medical Center, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Huanhuan Liu
- Department of Nuclear Medicine, The First Medical Center, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing 100853, China
- Graduate School, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Yanfeng Xu
- Nuclear Medicine Department, Beijing Friendship Hospital, Capital Medical University, 95 Yong'an Road, Xicheng District, Beijing 100050, China
| | - Wenwen Zhang
- Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Xiaohui Luan
- Department of Nuclear Medicine, The First Medical Center, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Xiaojie Liu
- Department of Nuclear Medicine, The First Medical Center, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Xiaodan Xu
- Department of Nuclear Medicine, The First Medical Center, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Shina Wu
- Department of Nuclear Medicine, The First Medical Center, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Guangyu Ma
- Department of Nuclear Medicine, The First Medical Center, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Ying Kan
- Nuclear Medicine Department, Beijing Friendship Hospital, Capital Medical University, 95 Yong'an Road, Xicheng District, Beijing 100050, China
| | - Jinming Zhang
- Department of Nuclear Medicine, The First Medical Center, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Ruimin Wang
- Department of Nuclear Medicine, The First Medical Center, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Jigang Yang
- Nuclear Medicine Department, Beijing Friendship Hospital, Capital Medical University, 95 Yong'an Road, Xicheng District, Beijing 100050, China
| |
Collapse
|
19
|
Wu H, Lv S, Zhang R, Gu L, Xu J, Li C, Zhang L, Shen F, Kow AWC, Wang M, Yang T. Next‐Generation Flexible Embolic Systems: Targeted Transarterial Chemoembolization Strategies for Hepatocellular Carcinoma. ADVANCED MATERIALS 2025. [DOI: 10.1002/adma.202503971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Indexed: 04/17/2025]
Abstract
AbstractTransarterial chemoembolization (TACE) remains the gold standard for treating intermediate‐stage hepatocellular carcinoma (HCC), yet faces great challenges in overcoming tumor heterogeneity, hypoxia‐induced angiogenesis, and metastatic progression. The development of advanced flexible embolization materials marks a revolutionary leap in interventional therapy, offering opportunities to revolutionize embolization precision, drug delivery kinetics, and tumor microenvironment modulation. This comprehensive review systematically examines the paradigm shift toward next‐generation TACE technology, emphasizing the limitations of conventional approaches and innovations in flexible embolic agents. A detailed discussion of next‐generation nano‐flexible embolic systems is presented, emphasizing their unique coagulation dynamics, real‐time imaging capabilities, and therapeutic precision. The review delves into groundbreaking TACE strategies integrating hypoxia modulation, energy conversion therapeutics, and sophisticated tumor microenvironment engineering. Clinical translation aspects are thoroughly explored, including large‐scale trial outcomes, vascular recanalization dynamics, and patient‐specific treatment optimization. Looking forward, key frontiers in the field is identified: intelligent nanocomposite systems, synergistic combination therapies, and precision medicine approaches tailored to individual tumor biology. This work not only objectively evaluates current progress but also charts future research priorities, aiming to transform TACE from a palliative intervention to a precision medicine platform and ultimately reshaping the landscape of HCC treatment and patient care.
Collapse
Affiliation(s)
- Han Wu
- Department of Hepatobiliary Surgery Eastern Hepatobiliary Surgery Hospital Naval Medical University Shanghai 200438 China
- Clinical research institute Eastern Hepatobiliary Surgery Hospital Naval Medical University Shanghai 200438 China
| | - Shaodong Lv
- Department of Hepatobiliary Surgery Eastern Hepatobiliary Surgery Hospital Naval Medical University Shanghai 200438 China
| | - Renjie Zhang
- School of Basic Medicine Naval Medical University Shanghai 200433 China
| | - Lihui Gu
- Department of Hepatobiliary Surgery Eastern Hepatobiliary Surgery Hospital Naval Medical University Shanghai 200438 China
| | - Jiahao Xu
- Department of Hepatobiliary Surgery Eastern Hepatobiliary Surgery Hospital Naval Medical University Shanghai 200438 China
| | - Chao Li
- Department of Hepatobiliary Surgery Eastern Hepatobiliary Surgery Hospital Naval Medical University Shanghai 200438 China
| | - Lijian Zhang
- School of Basic Medicine Naval Medical University Shanghai 200433 China
| | - Feng Shen
- Department of Hepatobiliary Surgery Eastern Hepatobiliary Surgery Hospital Naval Medical University Shanghai 200438 China
| | - Alfred Wei Chieh Kow
- Division of Hepatobiliary & Pancreatic Surgery Department of Surgery National University Hospital Singapore 119074 Singapore
| | - Mingda Wang
- Department of Hepatobiliary Surgery Eastern Hepatobiliary Surgery Hospital Naval Medical University Shanghai 200438 China
- Clinical research institute Eastern Hepatobiliary Surgery Hospital Naval Medical University Shanghai 200438 China
| | - Tian Yang
- Department of Hepatobiliary Surgery Eastern Hepatobiliary Surgery Hospital Naval Medical University Shanghai 200438 China
- Clinical research institute Eastern Hepatobiliary Surgery Hospital Naval Medical University Shanghai 200438 China
| |
Collapse
|
20
|
Jiang J, Zhang H, Ou Y, Lai J, Huang Y, Cai W, Li C, Zhang L, Fu Y. The immune-reinforcements of Lenvatinib plus anti-PD-1 and their rationale to unite with TACE for unresectable hepatocellular carcinoma treatment. Immunol Lett 2025; 275:107003. [PMID: 40189154 DOI: 10.1016/j.imlet.2025.107003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 03/05/2025] [Accepted: 03/26/2025] [Indexed: 04/12/2025]
Abstract
BACKGROUND Despite encouraging clinical benefits have gained by anti-PD-1 and Lenvatinib combination, in-depth characterizations about the mechanisms of action remain poorly characterized. Furthermore, although the combination of systemic anti-PD-1 or Lenvatinib treatment and locoregional transcatheter arterial chemoembolization (TACE) is widely carried out to treat unresectable HCC in clinical, the efficacies of different combination regimens are uncertain due to limited researches. METHODS We firstly generated murine HCC models to validate the enhanced anti-tumor effects of anti-PD-1 and Lenvatinib combination therapy. Then single cell mass cytometry (CyTOF) was employed to phenotypically reveal their mechanisms of action. After that, we further compared the effectiveness of TACE plus Lenvatinib (i.e., TACE-Len) dual therapy with TACE, Lenvatinib plus anti-PD-1 (i.e., TACE-Len-PD-1) triple therapy as conversion therapy for unresectable HCC. RESULTS Lenvatinib and anti-PD-1 combination could generate activated immune profiles not only by increasing systemic CD4+, CD8+T cells and B cells proportions, but also by weakening the immune-tolerance functions derived from both immunosuppressive cells (i.e., MDSCs) and co-inhibitory mediators (i.e., PD-L1 and LAG-3). Meanwhile, our study also suggested that TACE-Len-PD-1 triple therapy could achieve better clinical responses with powerful immune profiles for unresectable HCC compared to TACE-Len dual therapy. CONCLUSIONS Our study provided a delicate immune landscape of anti-PD-1and Lenvatinib combination, and we also offered scientific evidences that TACE-Len-PD-1 triple therapy could fulfill better clinical benefits than TACE-Len dual therapy, which is anticipated to provide objective and effective evidences for clinical use.
Collapse
Affiliation(s)
- Jiayun Jiang
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University/Army Medical University, Chongqing, 400038, PR China
| | - Hui Zhang
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University/Army Medical University, Chongqing, 400038, PR China
| | - Yanjiao Ou
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University/Army Medical University, Chongqing, 400038, PR China
| | - Jiejuan Lai
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University/Army Medical University, Chongqing, 400038, PR China
| | - Yulan Huang
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, PR China
| | - Wenyun Cai
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, PR China
| | - Chong Li
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, PR China.
| | - Leida Zhang
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University/Army Medical University, Chongqing, 400038, PR China.
| | - Yu Fu
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, PR China.
| |
Collapse
|
21
|
Zhang W, Hong X, Xiao Y, Wang H, Zeng X. Sorafenib resistance and therapeutic strategies in hepatocellular carcinoma. Biochim Biophys Acta Rev Cancer 2025; 1880:189310. [PMID: 40187502 DOI: 10.1016/j.bbcan.2025.189310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 03/30/2025] [Accepted: 03/30/2025] [Indexed: 04/07/2025]
Abstract
Hepatocellular carcinoma (HCC) remains one of the most prevalent and lethal cancers globally. While surgical resection and liver transplantation offer potential cures for early-stage HCC, the majority of patients are diagnosed at advanced stages where such interventions are not viable. Sorafenib, a multi-target kinase inhibitor, has been a cornerstone in the treatment of advanced HCC since its approval in 2007. Despite its significant clinical impact, less than half of the treated patients derive long-term benefits due to the emergence of resistance and associated side effects. This review focuses on the role of sorafenib, an FDA-approved multi-target kinase inhibitor, in treating advanced HCC, discusses the mechanisms underlying its therapeutic effects and associated resistance, and explores additional therapeutic strategies being investigated to improve patient outcomes.
Collapse
Affiliation(s)
- Weijing Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China
| | - Xuechuan Hong
- Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Yuling Xiao
- Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China; State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Hongbo Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China.
| | - Xiaodong Zeng
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China.
| |
Collapse
|
22
|
Xu J, Li J, Wang T, Luo X, Zhu Z, Wang Y, Wang Y, Zhang Z, Song R, Yang LZ, Wang H, Wong STC, Li H. Predicting treatment response and prognosis of immune checkpoint inhibitors-based combination therapy in advanced hepatocellular carcinoma using a longitudinal CT-based radiomics model: a multicenter study. BMC Cancer 2025; 25:602. [PMID: 40181337 PMCID: PMC11967134 DOI: 10.1186/s12885-025-13978-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 03/19/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Identifying effective predictive strategies to assess the response of immune checkpoint inhibitors (ICIs)-based combination therapy in advanced hepatocellular carcinoma (HCC) is crucial. This study presents a new longitudinal CT-based radiomics model to predict treatment response and prognosis in advanced HCC patients undergoing ICIs-based combination therapy. METHODS Longitudinal CT images were collected before and during the treatment for HCC patients across three institutions from January 2019 to April 2022. A total of 1316 radiomic features were extracted from arterial and portal venous phase abdominal CT images for each patient. A model called Longitudinal Whole-liver CT-based Radiomics (LWCTR) was developed to categorize patients into responders or non-responders using radiomic features and clinical information through support vector machine (SVM) classifiers. The area under the curve (AUC) was used as the performance metric and subsequently applied for risk stratification and prognostic assessment. The Shapley Additive explanations (SHAP) method was used to calculate the Shapley value, which explains the contribution of each feature in the SVM model to the prediction. RESULTS This study included 395 eligible participants, with a median age of 57 years (IQR 51-66), comprising 344 males and 51 females. The LWCTR model performed well in predicting treatment response, achieving an AUC of 0.883 (95% confidence interval [CI] 0.881-0.888) in the training cohort, 0.876 (0.858-0.895) in the internal validation cohort, and 0.875 (0.860-0.887) in the external test cohort. The Rad-Nomo model, integrating the LWCTR model's prediction score (Rad-score) with the modified Response Evaluation Criteria in Solid Tumors (mRECIST), demonstrated strong prognostic performance. It achieved time-dependent AUC values of 0.902, 0.823, and 0.850 at 1, 2, and 3 years in the internal validation cohort and 0.893, 0.848, and 0.762 at the same intervals in the external test cohort. CONCLUSION The proposed LWCTR model performs well in predicting treatment response and prognosis in patients with HCC receiving ICIs-based combination therapy, potentially contributing to personalized and timely treatment decisions.
Collapse
Affiliation(s)
- Jun Xu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China
- University of Science and Technology of China, Hefei, 230026, People's Republic of China
- Department of Intervention, The First Affiliated Hospital of University of Science and Technology of China, Hefei, 230001, People's Republic of China
- Department of Oncology, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China
| | - Junjun Li
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Hefei, 230001, People's Republic of China
| | - Tengfei Wang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China.
- University of Science and Technology of China, Hefei, 230026, People's Republic of China.
- Department of Oncology, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China.
| | - Xin Luo
- Yangtze Delta Region Institute (Huzhou) & School of Resources and Environment, University of Electronic Science and Technology of China, Huzhou, Chengdu, 313099, 611731, China
| | - Zhangxiang Zhu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Yimou Wang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China
- University of Science and Technology of China, Hefei, 230026, People's Republic of China
| | - Yong Wang
- Department of Radiology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, People's Republic of China
| | - Zhenglin Zhang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China
- University of Science and Technology of China, Hefei, 230026, People's Republic of China
| | - Ruipeng Song
- Department of Hepatobiliary Surgerydivision of Life Sciences and Medicineanhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, The First Affiliated Hospital of USTC, the University of Science and Technology of China, Hefei, 230001, People's Republic of China
| | - Li-Zhuang Yang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China
- University of Science and Technology of China, Hefei, 230026, People's Republic of China
- Department of Oncology, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China
| | - Hongzhi Wang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China
- University of Science and Technology of China, Hefei, 230026, People's Republic of China
- Department of Oncology, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China
| | - Stephen T C Wong
- Department of Systems Medicine and Bioengineering, Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX, 77030, USA
- Department of Radiology, Weill Cornell Medical College, New York, NY, 10065, United States
| | - Hai Li
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China.
- University of Science and Technology of China, Hefei, 230026, People's Republic of China.
- Department of Oncology, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China.
| |
Collapse
|
23
|
Tang HH, Zhang MQ, Zhang ZC, Fan C, Li SS, Chen W, Wang WD. Prognostic nutritional index predicts survival in intermediate and advanced hepatocellular carcinoma treated with hepatic arterial infusion chemotherapy combined with PD-(L)1 inhibitors and molecular targeted therapies. BMC Cancer 2025; 25:603. [PMID: 40181323 PMCID: PMC11966872 DOI: 10.1186/s12885-025-13993-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 03/21/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND This study aimed to evaluate the predictive efficacy of the prognostic nutritional index (PNI) in patients with intermediate and advanced hepatocellular carcinoma (HCC) treated with a regimen consisting of hepatic arterial infusion chemotherapy (HAIC), PD-(L)1 inhibitors, and molecular targeted therapies (MTTs). METHODS A retrospective analysis was performed on the data of 88 HCC patients received triple therapy between January 2020 and August 2022 at three medical centers. Univariate and multivariable analyses were conducted to assess the relationship between PNI and survival outcomes. RESULTS The median follow-up was 11.0 months (IQR: 8.0-17.0). The PNI cut-off value of 38.6 was determined using receiver operating characteristics (ROC) analysis. The median overall survival (OS) durations were 29.0 and 8.0 months in the high-PNI (≥ 38.6) and low-PNI (≤ 38.6) groups, respectively (HR = 0.306, 95% CI, 0.170-0.552, P < 0.001), and the median progression-free survival (PFS) durations were16.0 and 6.0 months, respectively (HR = 0.521, 95% CI, 0.303-0.896, P = 0.014). A higher complete response rate was observed in the high-PNI group (17.5% vs. 3.2%, P = 0.033). The univariate and multivariable analyses revealed that a PNI of ≥ 38.6 had an independent influence on both median OS (HR = 0.296; 95% CI, 0.159-0.551, P < 0.001) and median PFS (HR = 0.560; 95% CI, 0.318-0.987, P = 0.045). CONCLUSION The PNI is an objective and convenient tool that can potentially predict the prognosis of patients treated with HAIC-based triple therapy.
Collapse
Affiliation(s)
- Hao-Huan Tang
- Department of Interventional Radiology, Wuxi People's Hospital, Wuxi Medical Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Nanjing Medical University, No. 299, Wuxi, 214023, China
| | - Ming-Qing Zhang
- Department of Interventional Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Zi-Chen Zhang
- Department of Interventional Vascular Medicine, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, 230011, China
| | - Chen Fan
- Department of Interventional Radiology, Wuxi People's Hospital, Wuxi Medical Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Nanjing Medical University, No. 299, Wuxi, 214023, China
| | - Shu-Shu Li
- Department of Interventional Radiology, The Affiliated Wuxi No 5 People's Hospital of Jiangnan University, No. 1215, Guangrui Road, Wuxi, 214023, China
| | - Wei Chen
- Department of Interventional Radiology, The Affiliated Wuxi No 5 People's Hospital of Jiangnan University, No. 1215, Guangrui Road, Wuxi, 214023, China.
| | - Wei-Dong Wang
- Department of Interventional Radiology, Wuxi People's Hospital, Wuxi Medical Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Nanjing Medical University, No. 299, Wuxi, 214023, China.
| |
Collapse
|
24
|
Li J, Xian L, Wang X, Liu Y, Li J. The role of TACE in the era of immune-targeted therapy for hepatocellular carcinoma: a meta-analysis based on PSM. Front Immunol 2025; 16:1573834. [PMID: 40242754 PMCID: PMC12000099 DOI: 10.3389/fimmu.2025.1573834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 03/12/2025] [Indexed: 04/18/2025] Open
Abstract
Background Hepatocellular carcinoma (HCC) is a major global health challenge, with over 50% of patients ineligible for curative treatments at diagnosis. The combination of molecular targeted therapies and immunotherapy has shown promise in improving outcomes for advanced HCC. Objective This meta-analysis aims to assess the efficacy of combining transarterial chemoembolisation (TACE) with immune-targeted therapies in patients with unresectable HCC. Methods A systematic review and meta-analysis conforming to PRISMA guidelines were conducted by searching PubMed, Embase, Web of Science, and the Cochrane Library for studies published up to January 5, 2025. Due to the limited clinical evidence, our study exclusively included retrospective studies based on propensity score matching (PSM) analysis that compared the efficacy of TACE in combination with immune-targeted therapy to immune-targeted therapy alone. Key outcomes assessed included objective response rate (ORR), disease control rate (DCR), one-year overall survival (1-OS), one-year progression-free survival (1-PFS), median overall survival (mOS), and median progression-free survival (mPFS). Results A total of 9 PSM studies involving 2119 patients were included. The meta-analysis revealed that TACE significantly improved ORR, DCR, 1-OS, and 1-PFS, in addition to extending mOS and mPFS. Conclusion The findings suggest that the inclusion of TACE in treatment regimens for unresectable HCC notably enhances tumour control and patient survival. This study provides moderate to high-quality evidence supporting the integration of TACE in advanced HCC management, particularly for those patients not meeting standard TACE criteria. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/, identifier CRD 42025631817.
Collapse
Affiliation(s)
- Jiahao Li
- Department of Interventional Therapy, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Lei Xian
- Department of Interventional Therapy, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xinsen Wang
- Department of Interventional Therapy, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yingnan Liu
- Department of Radiology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Jiarui Li
- Department of Interventional Therapy, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
25
|
Ding G, Li K. A CT-Based Clinical-Radiomics Nomogram for Predicting the Overall Survival to TACE Combined with Camrelizumab and Apatinib in Patients with Advanced Hepatocellular Carcinoma. Acad Radiol 2025; 32:1993-2004. [PMID: 39578199 DOI: 10.1016/j.acra.2024.10.052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/16/2024] [Accepted: 10/30/2024] [Indexed: 11/24/2024]
Abstract
RATIONALE AND OBJECTIVES To construct a computed tomography (CT)-based clinical-radiomics nomogram for estimating overall survival (OS) in advanced hepatocellular carcinoma (HCC) patients receiving transcatheter arterial chemoembolization (TACE) in combination with camrelizumab and apatinib. METHODS A retrospective recruitment of 150 patients with clinically or pathologically confirmed HCC was conducted, followed by their division into training cohort (n = 105) and test cohort (n = 45). To generate the radiomics score (Rad-score), a series of analyses were performed, including Pearson correlation analysis, univariate Cox analysis, and least absolute shrinkage and selection operator Cox regression analysis. Subsequently, a clinical-radiomics nomogram was constructed using the Rad-score combined with independent clinical prognostic factors, followed by assessments of its calibration, discrimination, reclassification, and clinical utility. RESULTS Five CT radiomics features were selected. The Rad-score showed a significant correlation with OS (P < 0.001). The clinical-radiomics nomogram demonstrated superior performance in estimating OS, with a concordance index (C-index) of 0.840, compared to the radiomics nomogram (C-index: 0.817) and the clinical nomogram (C-index: 0.661). It also exhibited high 1-year and 2-year area under the curves of 0.936 and 0.946, respectively. Additionally, the clinical-radiomics nomogram markedly enhanced classification accuracy for OS outcomes, as evidenced by net reclassification improvement and integrated discrimination improvement. Decision curve analysis confirmed its clinical utility. CONCLUSION A CT-based clinical-radiomics nomogram exhibits strong potential for predicting OS in advanced HCC patients undergoing TACE combined with camrelizumab and apatinib.
Collapse
Affiliation(s)
- Guangyao Ding
- Department of General Surgery, Hefei BOE Hospital, Hefei, Anhui, China
| | - Kailang Li
- Department of General Surgery, Hefei BOE Hospital, Hefei, Anhui, China.
| |
Collapse
|
26
|
Zhong BY, Fan W, Guan JJ, Peng Z, Jia Z, Jin H, Jin ZC, Chen JJ, Zhu HD, Teng GJ. Combination locoregional and systemic therapies in hepatocellular carcinoma. Lancet Gastroenterol Hepatol 2025; 10:369-386. [PMID: 39993404 DOI: 10.1016/s2468-1253(24)00247-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 07/20/2024] [Accepted: 07/25/2024] [Indexed: 02/26/2025]
Abstract
Locoregional therapies play a fundamental role in the treatment of patients with early and intermediate and locally advanced hepatocellular carcinomas. With encouraging recent advances in immunotherapy-based systemic therapies, locoregional therapies are being both promoted and challenged by new systemic therapy options. Combined locoregional and systemic therapies might enhance treatment outcomes compared with either option alone. This Series paper summarises the existing data on locoregional and systemic therapies for hepatocellular carcinoma, and discusses evidence from studies investigating their combination with a focus on their synergistic efficacy and safety.
Collapse
Affiliation(s)
- Bin-Yan Zhong
- Center of Interventional Radiology and Vascular Surgery, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China; Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Wenzhe Fan
- Department of Interventional Oncology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Justin J Guan
- Division of Interventional Radiology, Department of Radiology, Cleveland Clinic, Cleveland, OH, USA
| | - Zhenwei Peng
- Department of Radiation Oncology, Cancer Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China; Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zhongzhi Jia
- Department of Interventional and Vascular Surgery, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
| | - Haojie Jin
- Shanghai Cancer Institute, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhi-Cheng Jin
- Center of Interventional Radiology and Vascular Surgery, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Jian-Jian Chen
- Center of Interventional Radiology and Vascular Surgery, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Hai-Dong Zhu
- Center of Interventional Radiology and Vascular Surgery, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Gao-Jun Teng
- Center of Interventional Radiology and Vascular Surgery, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China.
| |
Collapse
|
27
|
Xu Y, Xia D, Deng S, Liang M. Isoimperatorin Inhibits Angiogenesis by Suppressing VEGFR2 Signaling Pathway. Cardiovasc Drugs Ther 2025; 39:275-286. [PMID: 38363479 DOI: 10.1007/s10557-024-07561-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/07/2024] [Indexed: 02/17/2024]
Abstract
PURPOSE Angiogenesis involves in many pathological processes, including tumor metastasis, diabetic retinopathy, and rheumatoid arthritis. Therefore, identifying therapeutic drugs that target angiogenesis may be a promising strategy for disease treatment. Isoimperatorin is a furanocoumarin with anti-inflammatory and anti-microbial effects. However, the impacts of isoimperatorin on angiogenesis and its underlying mechanisms remain unclear. This study aimed to verify its effects on vascular endothelial growth factor (VEGF)-induced endothelial angiogenesis. METHODS We employed various assays including 5-ethynyl-2'-deoxyuridine incorporation assay, transwell migration assay, wound healing assay, tube formation assay, and Western blot to evaluate the effects of isoimperatorin on angiogenesis in vitro. Additionally, we utilized Western blot and immunofluorescence analysis to examine the activation of vascular endothelial growth factor receptor (VEGFR) 2 and its downstream signaling pathways following isoimperatorin treatment. To further validate the anti-angiogenic effects of isoimperatorin in vivo, we conducted a matrigel plug assay and established an orthotopic tumor model. RESULTS We demonstrated that pretreatment with isoimperatorin inhibited VEGF-induced endothelial cell proliferation, migration, and tube formation. Isoimperatorin also suppressed angiogenesis in vivo in a matrigel plug assay and in an orthotopic tumor model. Our results revealed that isoimperatorin exhibited anti-angiogenic effects via inhibiting VEGFR2 and its downstream signaling pathways activation. CONCLUSIONS Our study showed that isoimperatorin suppressed angiogenesis by targeting the VEGFR2 signaling pathway and could be a potential therapeutic agent for targeting angiogenesis.
Collapse
Affiliation(s)
- Yating Xu
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, China
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Di Xia
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, China
- Cardiovascular Center, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shan Deng
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, China.
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, China.
- Hubei clinical research center for metabolic and cardiovascular disease, Huazhong University of Science and Technology, Wuhan, China.
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Minglu Liang
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, China.
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, China.
- Hubei clinical research center for metabolic and cardiovascular disease, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
28
|
Liu L, Chen D, Wen L, Ma Y, Li J, Zhang G, Hu H, Huang C, Yao X. Efficacy and safety of fruquintinib combined with PD-1 inhibitors in the treatment of refractory metastatic colorectal cancer: a systematic review and meta-analysis. Expert Rev Anticancer Ther 2025; 25:411-421. [PMID: 40035688 DOI: 10.1080/14737140.2025.2474736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/01/2025] [Accepted: 02/23/2025] [Indexed: 03/06/2025]
Abstract
BACKGROUND Fruquintinib, a VEGFR1-3 tyrosine kinase inhibitor, is approved for treating refractory metastatic colorectal cancer. Recent clinical practice has shown that combining fruquintinib with programmed cell death protein 1 (PD-1) inhibitors can achieve better efficacy.The objective of this study is to assess the efficacy and safety of combining PD-1inhibitors with fruquintinib. METHODS We systematically searched PubMed, Cochrane Library, Embase, Wanfang, and CNKI up to 28 August 2024 for studies comparing fruquintinib combined with PD-1 inhibitors to fruquintinib alone. RevMan software was used to perform meta-analyses of survival data for the included studies. RESULTS A total of 9 retrospective cohort studies and 1 randomized controlled trial were included, involving a total of 716 patients. Compared with the monotherapy group, the combination therapy group had a greater Overall Response Rate [RR = 2.45,95% CI (1.83, 3.56), p < 0.00001], Disease Control Rate [RR = 1.37,95% CI (1.64,4.79), p = 0.0002], and progression-free survival [HR = 0.64,95% CI (0.49, 0.84), p = 0.001]. However, there was no significant difference in overall survival between the two groups. The incidence of adverse effects was identical in both groups. CONCLUSION Fruquintinib combined with PD-1 inhibitors was more effective than fruquintinib alone in the treatment of advanced colorectal cancer, with acceptable safety. REGISTRATION PROSPERO (registration number CRD42024583116).
Collapse
Affiliation(s)
- Linfeng Liu
- Gannan Medical University, Ganzhou, China
- Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Dengzhuo Chen
- Gannan Medical University, Ganzhou, China
- Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Liang Wen
- Gannan Medical University, Ganzhou, China
- Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yongli Ma
- Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jinghui Li
- Department of General Surgery, Xinfeng County People's Hospital, Xinfeng, Jiangxi, China
| | - Guosheng Zhang
- Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, China
| | - Hongkai Hu
- Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, China
| | - Chengzhi Huang
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xueqing Yao
- Gannan Medical University, Ganzhou, China
- Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| |
Collapse
|
29
|
Xu B, Wang LN, Wang ZY, He T, Zhu XD, Shen YH, Zhou J, Fan J, Sun HC, Huang C. Depth of Radiographic Response as an Independent Prognostic Factor for Patients with Initially Unresectable Hepatocellular Carcinoma Receiving Hepatectomy following Targeted Therapy plus Immunotherapy. Liver Cancer 2025; 14:142-157. [PMID: 40255871 PMCID: PMC12005705 DOI: 10.1159/000541300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 09/03/2024] [Indexed: 04/22/2025] Open
Abstract
Introduction Surgical resection following systemic therapy is feasible in patients with initially unresectable hepatocellular carcinoma (HCC). However, postoperative tumor recurrence is common after surgery, and the factors affecting this recurrence remain unclear. This study aimed to assess factors influencing postoperative outcomes in patients with initially unresectable HCC undergoing hepatectomy after systemic therapy. Methods This study retrospectively enrolled patients with initially unresectable HCC who underwent hepatectomy after targeted therapy plus immunotherapy (with or without locoregional therapy). Multivariate Cox regression analyses were used to identify the independent prognostic factors for recurrence-free survival (RFS) and overall survival (OS). Machine learning was used to determine the RFS rates at different intervals for different radiographic responses. Results Eighty-one patients who underwent R0 hepatectomy after systemic therapy were included. With a median follow-up of 17.4 (interquartile range: 7.2-22.3) months, median RFS and OS were not reached. Preoperative tumor downstaging and achieving pathological complete response were associated with improved RFS and OS. Multivariate Cox analyses identified radiographic response as an independent prognostic factor for RFS and OS. Furthermore, a radiographic response >40% (assessed using the Response Evaluation Criteria in Solid Tumors, version 1.1) or >50% (assessed using the modified Response Evaluation Criteria in Solid Tumors) was associated with a longer RFS (p = 0.006 and 0.003, respectively). Conclusion Radiographic response depth was an independent prognostic factor in patients with initially unresectable HCC who underwent hepatectomy following targeted therapy plus immunotherapy, and the response to systemic therapy may be the determining factor for patient prognosis after surgery.
Collapse
Affiliation(s)
- Bin Xu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lu-Na Wang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zi-Yi Wang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tian He
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiao-Dong Zhu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ying-Hao Shen
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jian Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jia Fan
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hui-Chuan Sun
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Cheng Huang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
30
|
TAN X, GU R, TAO J, ZHANG Y, SUN R, YIN G, ZHANG S, TANG D. Integrating network pharmacology and experimental validation to uncover the synergistic effects of Huangqi ()-Ezhu () with 5-fluorouracil in colorectal cancer models. J TRADIT CHIN MED 2025; 45:385-398. [PMID: 40151125 PMCID: PMC11955770 DOI: 10.19852/j.cnki.jtcm.2025.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 05/15/2024] [Indexed: 03/29/2025]
Abstract
OBJECTIVE To evaluate the effects of Huangqi (Radix Astragali Mongolici)-Ezhu (Rhizoma Curcumae Phaeocaulis) (HQEZ) on colorectal cancer therapies and to elucidate the potential mechanisms of HQEZ, especially in combination with 5-Fluorouracil (5-FU). METHODS The anti-tumor effects of HQEZ were evaluated in colorectal cancer models both in vivo and in vitro. The network pharmacological assay was used to investigate potential mechanisms of HQEZ. Potential target genes were selected by Gene Ontology (GO) enrichment analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis, protein-protein interaction network (PPI) and molecular docking. Within key targets, potential targets related to drug sensitivity, especially the sensitivity to 5-FU, were evaluated in HCT116 in vitro by immunofluorescence, quantitative real-time polymerase chain reaction (qPCR) and Western-blot. Then, changes in potential targets were assessed in tumors from tumor-bearing mice and the expression of these targets was also evaluated in colorectal cancer (COAD) patients from the Cancer Genome Atlas Program (TCGA) database. RESULTS HQEZ significantly enhanced the anti-tumor activity of 5-FU in vivo and inhibit the growth of HCT116 in vitro. By network pharmacological analysis, key targets, such as protein kinase B (AKT1), epidermal growth factor receptor (EGFR), adenosine triphosphate (ATP) binding cassette subfamily B member 1 (ABCB1, also named multidrug resistance protein 1, MDR1), ATP binding cassette subfamily G member 2 (ABCG2), thymidylate synthetase (TYMS, also named TS), prostaglandin-endoperoxide synthase 2 (PTGS2), matrix metallopeptidase 2 (MMP2), MMP9, toll like receptor 4 (TLR4), TLR9 and dihydropyrimidine dehydrogenase (DPYD), were identified. Additionally, 4 potential core active ingredients (Folate, Curcumin, quercetin and kaempferol) were identified to be important for the treatment of colorectal cancer with HQEZ. In key targets, chemoresistance related targets were validated to be affected by HQEZ. Furthermore, 5-FU sensitivity related targets, including MDR1, TS, EGFR, ribonucleotide reductase catalytic subunit M1, Breast and Ovarian Cancer Susceptibility Protein 1 (BRCA1) and mutl homolog 1 were also significantly reduced by HQEZ both in vitro and in vivo. Finally, these validated key targets and 5-FU sensitivity related targets were demonstrated to be up-regulated in COAD patients based on TCGA database. CONCLUSION HQEZ has synergistic effects on the anti-tumor activity of 5-FU in the treatment of colorectal cancer both in vivo and in vitro. The beneficial effect of HQEZ results from the inhibition of the drug sensitivity targets associated with 5-FU. The combination therapy of HQEZ with 5-FU or other chemotherapeutic drugs will also improve the anti-tumor efficacy of chemotherapy.
Collapse
Affiliation(s)
- Xiying TAN
- 1 Department of Pharmacy, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Ruxin GU
- 2 Department of Pain Management, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, China
- 3 School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Jing TAO
- 4 School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yu ZHANG
- 3 School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - RuiQian SUN
- 5 School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Gang YIN
- 5 School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Shuo ZHANG
- 6 Department of Pharmacy, Nantong Hospital of Traditional Chinese Medicine, Nantong, 226001, China
| | - Decai TANG
- 5 School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
31
|
He K, Xiao Y, Tu S, Li Y, Wu Z, Liu L, Shen W, Bao S, He Y. Efficacy evaluation of postoperative adjuvant transarterial chemoembolization in preventing hepatocellular carcinoma recurrence within the Milan criteria: A multicenter propensity score matching analysis based on pathologic indicators. J Gastrointest Surg 2025; 29:101978. [PMID: 39900235 DOI: 10.1016/j.gassur.2025.101978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 01/13/2025] [Accepted: 01/25/2025] [Indexed: 02/05/2025]
Abstract
BACKGROUND Malignant biologic behaviors, such as microvascular invasion (MVI), satellite nodule formation, and poor differentiation, can appear in the postoperative pathology of early hepatocellular carcinoma (HCC), which often indicates an earlier stage of malignant evolution. This study aimed to evaluate tumor recurrence in patients with HCC meeting the Milan criteria who underwent postoperative adjuvant transarterial chemoembolization (PA-TACE) according to postoperative pathologic indices. METHODS A retrospective study was conducted on 790 patients with HCC meeting the Milan criteria who underwent hepatectomy across 4 medical centers, consisting of 366 patients treated with PA-TACE and 424 patients treated without PA-TACE. To reduce selection bias, propensity score matching (PSM) at a 1:1 ratio was applied, achieving balanced clinical characteristics between the 2 groups. RESULTS Patients who underwent PA-TACE did not experience severe adverse events or toxicity-related deaths. After PSM of each subgroup, it was found that patients with MVI (median time: 37 vs 17 months; P =.010), satellite nodules (median time: Not Applicable [NA] vs 14 months; P =.018), and Edmondson-Steiner grades III and IV (median time: NA vs 13 months; P =.004) who received PA-TACE had higher recurrence-free survival (RFS). However, patients who were MVI negative, satellite nodule negative, and Edmondson-Steiner grades I and II did not benefit from PA-TACE in terms of RFS (all P >.05). Patients who received PA-TACE were more likely to undergo liver transplantation, rehepatectomy, or local ablation after tumor recurrence, whereas those who did not receive PA-TACE were more likely to receive TACE, chemoradiotherapy, or immune-targeted therapy after tumor recurrence (all P<.05). CONCLUSION Postoperative pathologic indicators can guide the selection of PA-TACE for patients with HCC meeting the Milan criteria. Patients with MVI, satellite nodules, and Edmondson-Steiner grades III and IV are more suitable for receiving PA-TACE to improve RFS. PA-TACE may alter the recurrence pattern of tumors, rendering them more localized.
Collapse
Affiliation(s)
- Kun He
- Department of Hepatobiliary Surgery, Zhongshan People's Hospital, Zhongshan Hospital Affiliated to Sun Yat-sen University, Zhongshan City, China
| | - Yongqiang Xiao
- Department of General Surgery, Ganjiang New Area People's Hospital, Ganjiang New Area Hospital of the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Shuju Tu
- Department of Hepatobiliary Surgery, Xiantao First People's Hospital, Xiantao City, China
| | - Youyao Li
- Division of Hepatobiliary and Pancreas Surgery, Department of General Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University and The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen City, China
| | - Zhao Wu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Liping Liu
- Division of Hepatobiliary and Pancreas Surgery, Department of General Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University and The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen City, China
| | - Wei Shen
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Shiyun Bao
- Division of Hepatobiliary and Pancreas Surgery, Department of General Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University and The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen City, China
| | - Yongzhu He
- Division of Hepatobiliary and Pancreas Surgery, Department of General Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University and The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen City, China.
| |
Collapse
|
32
|
Kang W, Tang P, Luo Y, Lian Q, Zhou X, Ren J, Cong T, Miao L, Li H, Huang X, Ou A, Li H, Yan Z, Di Y, Li X, Ye F, Zhu X, Yang Z. Multiparametric MRI-based Machine Learning Radiomics for Predicting Treatment Response to Transarterial Chemoembolization Combined with Targeted and Immunotherapy in Unresectable Hepatocellular Carcinoma: A Multicenter Study. Acad Radiol 2025; 32:2013-2026. [PMID: 39609145 DOI: 10.1016/j.acra.2024.10.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/30/2024]
Abstract
RATIONALE AND OBJECTIVES To develop and validate multiple machine learning predictive models incorporating clinical features and pretreatment multiparametric magnetic resonance imaging (MRI) radiomic features for predicting treatment response to transarterial chemoembolization combined with molecular targeted therapy plus immunotherapy in unresectable hepatocellular carcinoma (HCC). MATERIALS AND METHODS This retrospective study involved 276 patients with unresectable HCC who received combination therapy from 4 medical centers. Patients were divided into one training cohort and two independent external validation cohorts. 16 radiomic features from six multiparametric MRI sequences and 2 clinical features were used to build six machine learning models. The models were evaluated using the area under the curve (AUC), decision curve analysis, and incremental predictive value. RESULTS Alpha-fetoprotein and neutrophil-to-lymphocyte ratio are clinical independent predictors of treatment response. In the training cohort and two external validation cohorts, the AUCs and 95% confidence intervals for predicting treatment response were respectively 0.782 (0.698-0.857) 0.695 (0.566-0.823), and 0.679 (0.542-0.810) for the clinical model; 0.942 (0.903-0.974), 0.869 (0.761-0.949), and 0.868 (0.769-0.942) for the radiomics model; and 0.956 (0.920-0.984), 0.895 (0.810-0.967), and 0.892 (0.804-0.957) for the combined clinical-radiomics model. In the three cohorts, the incremental predictive value of the radiomics model over the clinical model was 49.2% (P < 0.001), 28.8% (P < 0.001), and 31.5% (P < 0.001). CONCLUSION The combined clinical-radiomics model may provide a reliable and non-invasive tool to predict individual treatment responses and guide and improve clinical decision-making in combination therapy of HCC patients.
Collapse
Affiliation(s)
- Wendi Kang
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Peiyun Tang
- Department of Radiology, China-Japan Union Hospital of Jilin University, Changchun 130000, China
| | - Yingen Luo
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Qicai Lian
- Department of Interventional Radiology, the Affiliated Cancer Hospital of Guizhou Medical University, Guiyang 550000, China
| | - Xuan Zhou
- Department of Radiology, Third Xiangya Hospital, Central South University, Changsha, 410013 Hunan, China
| | - Jinrui Ren
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Tianhao Cong
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Lei Miao
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Hang Li
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xiaoyu Huang
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Aixin Ou
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Hao Li
- Department of Interventional Radiology, The First Hospital of China Medical University, No.155 Nanjing Road, Heping District, Shenyang 110001, Liaoning, China
| | - Zhentao Yan
- Department of Interventional Radiology, The First Hospital of China Medical University, No.155 Nanjing Road, Heping District, Shenyang 110001, Liaoning, China
| | - Yingjie Di
- Department of Interventional Therapy, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan 030013, China
| | - Xiao Li
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Feng Ye
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xiaoli Zhu
- Department of Interventional Radiology, The First Affiliated Hospital, Soochow University, No.188 Shizi Road, Suzhou 215006, China
| | - Zhengqiang Yang
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| |
Collapse
|
33
|
Yang J, Zhang S, Yin Y, Li Z, Lu S, Ni G, Zhu X, Ni C. Efficacy and safety of hepatic arterial infusion chemotherapy followed by transarterial embolization for hepatocellular carcinoma. TUMORI JOURNAL 2025; 111:158-163. [PMID: 40103179 DOI: 10.1177/03008916251327830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
PURPOSE To compare the safety and efficacy of hepatic arterial infusion chemotherapy followed by transarterial embolization (HAIC+TAE) to transarterial chemoembolization (TACE) for the treatment of unresectable hepatocellular carcinoma (uHCC). MATERIALS AND METHODS The clinical data of patients who received HAIC+TAE or TACE between April 2020 and April 2022 was collected. Propensity score-matching was used to balance the baseline characteristics of the two groups. Tumor response according to mRECIST, median time to progression (TTP) and overall survival (OS) were investigated. ALBI score was applied to evaluate the changes of liver function and other relative adverse reactions were recorded. RESULTS A total of 98 patients with uHCC were enrolled in the study, including 71 in the TACE group and 27 in the HAIC+TAE group. After propensity score matching, 23 pairs of patients were investigated. The HAIC+TAE group showed a longer median TTP and OS than TACE group (mTTP 316 vs. 235 days, P=0.023; mOS 580 vs. 493 days, P=0.020). Objective response rates in HAIC+TAE group and TACE group were 65.2% and 47.8% (P=0.234). Disease-control rates were 87.0% and 82.6% (P=1.000). No significant difference was found in the incidence of adverse events between the two groups (P>0.05). CONCLUSION The combination treatment strategy of HAIC+TAE in patients with uHCC appears to be a safe regimen, with the potential to prolong mTTP and mOS relative to TACE. The sequential application of this therapy merits consideration as an innovative treatment strategy for individuals with uHCC.
Collapse
Affiliation(s)
- Jun Yang
- Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Shen Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yu Yin
- Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhi Li
- Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Shitao Lu
- Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Guanyin Ni
- Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaoli Zhu
- Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Caifang Ni
- Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
34
|
Liu L, Cai Q, Wu D, Li S, Liu D, Zheng J. AEBP1 inhibition reduces cell growth and PI3K/AKT pathway while less regulates cell mobility in hepatocellular carcinoma. World J Surg Oncol 2025; 23:108. [PMID: 40158165 PMCID: PMC11954352 DOI: 10.1186/s12957-025-03750-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 03/08/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Adipocyte enhancer-binding protein 1 (AEBP1) regulates collagen fibrosis, extracellular matrix, and important oncogene pathways, but its regulation on hepatocellular carcinoma (HCC) is not known. This study aimed to investigate the effect of AEBP1 knockdown on HCC cell proliferation, apoptosis, migration, invasion and PI3K/AKT pathway. METHODS MHCC-97 H and Huh7 cell lines were applied. Negative control or AEBP1 siRNA (siAEBP1) were transfected into cells, and cells without transfection were set as blank control. Quantitative polymerase chain reaction (qPCR), western blot, Cell Counting Kit-8 (CCK-8), 5-Ethynyl-2'-deoxyuridine (EdU) staining, Terminal-deoxynucleotidyl Transferase Mediated Nick End Labeling (TUNEL) staining, Transwell invasion, and cell scratch assays were performed. RESULTS AEBP1 mRNA and protein expressions were lower after siAEBP1 transfection in MHCC-97 H and Huh7 cells. OD value of CCK-8 and EdU positive cell percentage were decreased, while TUNEL reflected cell apoptosis rate was increased, after siAEBP1 transfection in MHCC-97 H and Huh7 cells. However, invasive cell number and cell migration rate were only reduced after siAEBP1 transfection in Huh7 cells but not in MHCC-97 H cells. Expressions of p-PI3K/PI3K and p-AKT/AKT were downregulated after siAEBP1 transfection in MHCC-97 H and Huh7 cells. Subsequent rescue experiment revealed that th activation of PI3K/AKT pathway by 740Y-P attenuated the effect of siAEBP1 transfection in MHCC-97 H and Huh7 cells. CONCLUSION AEBP1 exhibits the potency to be a target for HCC treatment, reflected by its regulation on HCC proliferation, apoptosis and PI3K/AKT pathway, but its effect on HCC invasion and migration seems limited.
Collapse
Affiliation(s)
- Liyou Liu
- Department of Hepatobiliary Surgery, Tangshan Central Hospital, No. 601-1 Changning Road, Lubei District, Tangshan, 064000, Hebei Province, China
| | - Qingshan Cai
- Department of Hepatobiliary Surgery, Tangshan Central Hospital, No. 601-1 Changning Road, Lubei District, Tangshan, 064000, Hebei Province, China
| | - Dongyang Wu
- Department of Hepatobiliary Surgery, Tangshan Central Hospital, No. 601-1 Changning Road, Lubei District, Tangshan, 064000, Hebei Province, China
| | - Shudong Li
- Department of Hepatobiliary Surgery, Tangshan Central Hospital, No. 601-1 Changning Road, Lubei District, Tangshan, 064000, Hebei Province, China
| | - Dong Liu
- Department of Hepatobiliary Surgery, Tangshan Central Hospital, No. 601-1 Changning Road, Lubei District, Tangshan, 064000, Hebei Province, China
| | - Jianxing Zheng
- Department of Hepatobiliary Surgery, Tangshan Central Hospital, No. 601-1 Changning Road, Lubei District, Tangshan, 064000, Hebei Province, China.
| |
Collapse
|
35
|
Zhang XD, Zhang LY, Luo JL, Yu KH, Zhu KL. Neoadjuvant therapy: Dawn of reducing the high post-surgery recurrence rate of hepatocellular carcinoma. World J Gastrointest Surg 2025; 17:103740. [PMID: 40162404 PMCID: PMC11948117 DOI: 10.4240/wjgs.v17.i3.103740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/01/2025] [Accepted: 01/14/2025] [Indexed: 02/24/2025] Open
Abstract
The high postoperative recurrence rate remains a major challenge in the treatment of hepatocellular carcinoma (HCC) following resection. Increasing research has been delved into investigating the role of neoadjuvant therapy on the prognosis of resectable HCC. Recent trends in combination therapy with molecularly targeted agents and immune checkpoint inhibitors have significantly improved the efficacy of systemic antitumor treatments, yielding survival benefits exceeding 40%. Neoadjuvant therapy for HCC, whether based on systemic antitumor treatments, locoregional therapies, or their combination, has emerged as a promising research direction. However, there remains a matter of debate on neoadjuvant therapy. In this review, we summarize and discuss the research progress and challenges of neoadjuvant therapy for HCC over the past five years from the perspective of Chinese guidelines to provide new insights and future directions in this field.
Collapse
Affiliation(s)
- Xiao-Dong Zhang
- Department of Hepatopancreatobiliary Surgery, The Affiliated People's Hospital of Ningbo University, Ningbo 315040, Zhejiang Province, China
| | - Lu-Yi Zhang
- Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo 315211, Zhejiang Province, China
| | - Jia-Liang Luo
- Department of Hepatopancreatobiliary Surgery, The Affiliated People's Hospital of Ningbo University, Ningbo 315040, Zhejiang Province, China
| | - Ke-Heng Yu
- Department of Hepatopancreatobiliary Surgery, The Affiliated People's Hospital of Ningbo University, Ningbo 315040, Zhejiang Province, China
| | - Ke-Lei Zhu
- Department of Hepatopancreatobiliary Surgery, The Affiliated People's Hospital of Ningbo University, Ningbo 315040, Zhejiang Province, China
| |
Collapse
|
36
|
Liu S, Wu H, Zhang P, Zhou H, Wu D, Jin Y, Yang H, Xing R, Wu Y, Wu G. NELL2 suppresses epithelial-mesenchymal transition and induces ferroptosis via notch signaling pathway in HCC. Sci Rep 2025; 15:10193. [PMID: 40133552 PMCID: PMC11937300 DOI: 10.1038/s41598-025-94669-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 03/17/2025] [Indexed: 03/27/2025] Open
Abstract
Although various malignant tumors have been associated with the aberrant expression of Neural Epidermal Growth Factor-Like 2 (NELL2), its involvement in hepatocellular carcinoma (HCC) has not been previously documented. In this study, NELL2, recognized as a crucial tumor-suppressor gene, was found to be infrequently expressed in HCC. In vitro experiments demonstrated that the overexpression of NELL2 significantly inhibited the proliferation, migration, and invasion of liver cancer cells, whereas the suppression of NELL2 markedly enhanced these oncogenic properties. Further investigation revealed that NELL2 impedes epithelial-mesenchymal transition (EMT) via the Notch signaling pathway. Inhibition of the Notch pathway reversed the increased tumor proliferation, migration, and invasion observed following the downregulation of NELL2 expression. Notably, gene enrichment analysis and in vitro studies indicated that NELL2 effectively induced ferroptosis in HCC cells, as evidenced by increased levels of cellular malondialdehyde (MDA), iron, and Reactive Oxygen Species (ROS), alongside decreased glutathione (GSH) levels. The blockade of the Notch signaling pathway substantially diminished NELL2's capacity to induce ferroptosis. In summary, our findings suggest that NELL2 modulates the Notch signaling pathway to inhibit EMT and promote ferroptosis. Consequently, NELL2 may serve as a novel therapeutic target, potentially functioning as a tumor suppressor gene in HCC.
Collapse
Affiliation(s)
- Shiqi Liu
- Hepatobiliary Surgery Department, First Hospital of China Medical University, No.155, Nanjingbei Street, Shenyang, 110001, Liaoning, People's Republic of China
- Key Laboratory of General Surgery of Liaoning Province, First Hospital of China Medical University, No.155, Nanjingbei Street, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Haomin Wu
- Hepatobiliary Surgery Department, First Hospital of China Medical University, No.155, Nanjingbei Street, Shenyang, 110001, Liaoning, People's Republic of China
- Key Laboratory of General Surgery of Liaoning Province, First Hospital of China Medical University, No.155, Nanjingbei Street, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Pengjie Zhang
- Hepatobiliary Surgery Department, First Hospital of China Medical University, No.155, Nanjingbei Street, Shenyang, 110001, Liaoning, People's Republic of China
| | - Haonan Zhou
- Hepatobiliary Surgery Department, First Hospital of China Medical University, No.155, Nanjingbei Street, Shenyang, 110001, Liaoning, People's Republic of China
| | - Di Wu
- Hepatobiliary Surgery Department, First Hospital of China Medical University, No.155, Nanjingbei Street, Shenyang, 110001, Liaoning, People's Republic of China
| | - Yifan Jin
- Hepatobiliary Surgery Department, First Hospital of China Medical University, No.155, Nanjingbei Street, Shenyang, 110001, Liaoning, People's Republic of China
- Key Laboratory of General Surgery of Liaoning Province, First Hospital of China Medical University, No.155, Nanjingbei Street, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Hongwei Yang
- Hepatobiliary Surgery Department, First Hospital of China Medical University, No.155, Nanjingbei Street, Shenyang, 110001, Liaoning, People's Republic of China
- Key Laboratory of General Surgery of Liaoning Province, First Hospital of China Medical University, No.155, Nanjingbei Street, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Ruilin Xing
- Hepatobiliary Surgery Department, First Hospital of China Medical University, No.155, Nanjingbei Street, Shenyang, 110001, Liaoning, People's Republic of China
| | - Yubo Wu
- Hepatobiliary Surgery Department, First Hospital of China Medical University, No.155, Nanjingbei Street, Shenyang, 110001, Liaoning, People's Republic of China
| | - Gang Wu
- Hepatobiliary Surgery Department, First Hospital of China Medical University, No.155, Nanjingbei Street, Shenyang, 110001, Liaoning, People's Republic of China.
- Key Laboratory of General Surgery of Liaoning Province, First Hospital of China Medical University, No.155, Nanjingbei Street, Shenyang, 110001, Liaoning Province, People's Republic of China.
| |
Collapse
|
37
|
Messaoudi N, Vanlander A, Benhadda M, Makarian R, Kortbeek K, De Haar-Holleman A, Gumbs AA. Hepatic arterial infusion pump chemotherapy for colorectal liver metastases: Revisiting traditional techniques to explore new frontiers. World J Clin Oncol 2025; 16:101274. [PMID: 40130052 PMCID: PMC11866082 DOI: 10.5306/wjco.v16.i3.101274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/14/2024] [Accepted: 12/09/2024] [Indexed: 01/21/2025] Open
Abstract
Hepatic arterial infusion (HAI) chemotherapy, first introduced in the 1980s, has gained recognition as an effective locoregional treatment for colorectal liver metastasis (CRLM). Initially used for unresectable liver metastases, HAI's application has expanded to the adjuvant setting following hepatic resection, with early studies indicating improved hepatic disease-free survival. Recent research demonstrates that combining HAI with modern systemic therapies enhances conversion to resectability and prolongs both recurrence-free and overall survival, even in heavily pretreated patients with diverse RAS mutational statuses. Personalization through approaches like microsatellite instability status and dose modifications further optimize outcomes. However, the complexity of HAI requires expertise across multidisciplinary teams, limiting its widespread adoption to specialized centers. Ongoing clinical trials continue to investigate HAI's role in CRLM management, highlighting its potential to become a cornerstone of liver-directed therapy. We explore how HAI chemotherapy, in combination with personalized medicine, can advance treatment strategies for metastatic colorectal cancer.
Collapse
Affiliation(s)
- Nouredin Messaoudi
- Department of Hepatopancreatobiliary Surgery, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel and Europe Hospitals, Brussels 1090, Brussels-Capital Region, Belgium
| | - Aude Vanlander
- Department of Hepatopancreatobiliary Surgery, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel and Europe Hospitals, Brussels 1090, Brussels-Capital Region, Belgium
| | - Myriam Benhadda
- Department of Hepatopancreatobiliary Surgery, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel and Europe Hospitals, Brussels 1090, Brussels-Capital Region, Belgium
| | - Roza Makarian
- Department of Hepatopancreatobiliary Surgery, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel and Europe Hospitals, Brussels 1090, Brussels-Capital Region, Belgium
| | - Koen Kortbeek
- Department of Oncology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels 1090, Brussels-Capital Region, Belgium
| | - Amy De Haar-Holleman
- Department of Oncology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels 1090, Brussels-Capital Region, Belgium
| | - Andrew A Gumbs
- Service de Chirurgie Digestive Minimale Invasive, Hôpital Antoine Béclère, Assistance Publique-Hôpitaux de Paris, Clamart 92140, France
- Department of Surgery, University of Magdeburg, Magdeburg 39130, Saxony-Anhalt, Germany
| |
Collapse
|
38
|
Liang Y, Ruan T, He J, Huang K, Wei M, Tan S. Long-term survival in a patient with ruptured advanced hepatocellular carcinoma treated with nutritional therapy combined with apatinib and camrelizumab: a case report. Discov Oncol 2025; 16:378. [PMID: 40121613 PMCID: PMC11930898 DOI: 10.1007/s12672-025-02099-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 03/10/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Spontaneous rupture is a fatal complication of advanced hepatocellular carcinoma (HCC) with an extremely poor prognosis. Although immune checkpoint inhibitors, targeted therapies, and nutritional therapy have shown potential in the treatment of advanced HCC, their combined efficacy in complex cases with high tumor burden complicated by rupture and bleeding remains unclear. CASE DESCRIPTION A 54-year-old male patient was diagnosed with Barcelona Clinic Liver Cancer (BCLC) stage C HCC with high tumor burden, accompanied by a history of chronic hepatitis B and moderate malnutrition. After initial treatment with apatinib (500 mg/day) and nutritional therapy, the patient experienced HCC rupture. Following emergency transarterial embolization for hemostasis, the treatment regimen was adjusted to camrelizumab (200 mg/2 weeks) combined with reduced-dose apatinib (250 mg/day), along with continued nutritional support. After 17 months of treatment, the patient underwent hepatectomy, with pathological examination showing complete remission in the left liver. Postoperative adjuvant therapy included transarterial chemoembolization, nutritional therapy, targeted therapy, and individualized immunotherapy. As of the 4-year follow-up, the patients has good quality of life and has not experienced recurrence. CONCLUSION This case showcases a multimodal treatment strategy for patients with advanced HCC with high tumor burden and rupture complications, integrating individualized immuno-targeted therapy, interventional treatment, and nutritional management and providing a possible approach for achieving a long-term survival. This comprehensive treatment method may offer new insights into improving the prognosis of patients with advanced HCC.
Collapse
Affiliation(s)
- Yaohao Liang
- Department of Hepatobiliary and Pancreatic Surgery, Nanning Ninth People's Hospital, #233 Yong'an West Road, Nanning, 530400, Guangxi, China
- Department of Hepatobiliary and Pancreatic Surgery, Liuzhou People's Hospital Affiliated to Guangxi Medical University, Liuzhou, 545006, Guangxi, China
| | - Tianyu Ruan
- Department of Radiology, Liuzhou People's Hospital Affiliated to Guangxi Medical University, Liuzhou, 545006, Guangxi, China
| | - Jiaqian He
- Department of Radiology, Liuzhou People's Hospital Affiliated to Guangxi Medical University, Liuzhou, 545006, Guangxi, China
| | - Ketuan Huang
- Department of Hepatobiliary and Pancreatic Surgery, Liuzhou People's Hospital Affiliated to Guangxi Medical University, Liuzhou, 545006, Guangxi, China
| | - Min Wei
- Department of Hepatobiliary and Pancreatic Surgery, Liuzhou People's Hospital Affiliated to Guangxi Medical University, Liuzhou, 545006, Guangxi, China
| | - Shengqiang Tan
- Department of Hepatobiliary and Pancreatic Surgery, Nanning Ninth People's Hospital, #233 Yong'an West Road, Nanning, 530400, Guangxi, China.
| |
Collapse
|
39
|
Zhang X, Wang B, Qi X, Qian Z, Gao X, Cheng Y, Wang X. A Glutathione-Responsive System with Prodrug and Sensitization Strategies for Targeted Therapy of Glioma. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2501620. [PMID: 40119786 DOI: 10.1002/smll.202501620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 03/08/2025] [Indexed: 03/24/2025]
Abstract
Glioblastoma represents a highly aggressive form of malignant tumor within the central nervous system. Although chemotherapy remains the primary therapeutic strategy, its efficacy is often limited. To overcome the limitations associated with chemotherapeutic agents, such as high toxicity and non-specific adverse effects, a novel nanoparticle system comprising cRGD-modified and glutathione (GSH)-responsive polymers, and PEG-ss-Dox and apatinib (AP) (PDOX-AP/cRGD-NPs) is developed. PDOX-AP/cRGD-NPs show effective penetration of the blood-brain barrier (BBB), facilitate targeted delivery to brain tumors, and exhibit controlled drug release. PDOX-AP/cRGD-NPs show more effect in reducing the viability of GL-261, U87-MG, and LN-229 cells, inhibiting clonogenicity, and suppressing anti-apoptotic protein expression than PDOX/cRGD-NPs or AP/cRGD-NPs. Additionally, PDOX-AP/cRGD-NPs substantially increase drug uptake, BBB penetration, apoptosis rates, and the proportion of cells in the G2 phase. In vivo experiments further reveal that cRGD-directed nanoparticles exhibit superior accumulation in glioma regions compared to their non-cRGD-modified counterparts. In the interim, PDOX-AP/cRGD-NPs demonstrate significant efficacy in suppressing both ectopic and orthotopic growth of GL-261 gliomas, as well as orthotopic LN-229 gliomas, thereby markedly extending the median survival duration. This study introduces a promising targeted co-delivery system for combination chemotherapy.
Collapse
Affiliation(s)
- Xifeng Zhang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Bilan Wang
- Department of Pharmacy, West China Second University Hospital of Sichuan University, Chengdu, 610041, P. R. China
| | - Xin Qi
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Zhiyong Qian
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Yongzhong Cheng
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Xiang Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| |
Collapse
|
40
|
Wu Y, Xie BB, Zhang BL, Zhuang QX, Liu SW, Pan HM. Apatinib regulates the glycolysis of vascular endothelial cells through PI3K/AKT/PFKFB3 pathway in hepatocellular carcinoma. World J Gastroenterol 2025; 31:102848. [PMID: 40124275 PMCID: PMC11924011 DOI: 10.3748/wjg.v31.i11.102848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/17/2025] [Accepted: 02/11/2025] [Indexed: 03/13/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a prevalent and aggressive malignancy in the Chinese population; the severe vascularization by the tumor makes it difficult to cure. The high incidence and poor survival rates of this disease indicate the search for new therapeutic alternatives. Apatinib became a drug of choice because it inhibits tyrosine kinase activity, mainly through an effect on vascular endothelial growth factor receptor-2, thereby preventing tumor angiogenesis. This mechanism of action makes apatinib effective in the treatment of HCC. AIM To investigate the effect of apatinib on the glycolysis of vascular endothelial cells (VECs). METHODS This present study has investigated the effects of HCC cells on VECs, paying particular attention to changes in the glycolytic activity of VECs. The co-culture system established in the present study examined key cellular functions such as extracellular acidification rate and oxygen consumption rate. It also discusses participation of apatinib in the above processes. Core to the findings is the phosphatidylinositol 3-kinase (PI3K)/AKT/6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) signaling pathway, emphasizing the function of phosphorylated AKT and its interaction with PFKFB3, an essential regulator of glycolysis. In the investigation, molecular mechanisms by which such a pathway could influence the above VECs functions of proliferation, migration, and tube formation were underlined through coimmunoprecipitation analysis. Besides, supplementary in vivo experiments on nude mice provided additional biological relevance to the obtained results. RESULTS The glycolytic metabolism in VECs co-cultured with HCC cells is highly active, and the increased glycolysis in these endothelial cells accelerates the malignant transformation of HCC cells. Apatinib has been shown to inhibit this glycolytic activity in the VECs. It also hinders the development, multiplication, and movement of these cells while encouraging their programmed cell death. Moreover, biological analysis revealed that apatinib mainly influences VECs by regulating the PI3K/AKT signaling pathway. Subsequent research indicated that apatinib blocks the PI3K/AKT/PFKEB3 pathway, which in turn reduces glycolysis in these cells. CONCLUSION Apatinib influences the glycolytic pathway in the VECs of HCC a through the PI3K/AKT/PFKFB3 signaling pathway.
Collapse
Affiliation(s)
- Yi Wu
- Division of Cancer Medicine, Sir Run Run Shaw Medical Center, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
| | - Bin-Bin Xie
- Division of Cancer Medicine, Sir Run Run Shaw Medical Center, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
| | - Bing-Liang Zhang
- Section of Oncology, Ningxia Hui Autonomous Region General Hospital, Ningxia Medical University, Yinchuan 750000, Ningxia Hui Autonomous Region, China
| | - Qing-Xin Zhuang
- Section of Oncology, Ningxia Hui Autonomous Region General Hospital, Ningxia Medical University, Yinchuan 750000, Ningxia Hui Autonomous Region, China
| | - Shi-Wei Liu
- Section of Oncology, Ningxia Hui Autonomous Region General Hospital, Ningxia Medical University, Yinchuan 750000, Ningxia Hui Autonomous Region, China
| | - Hong-Ming Pan
- Division of Cancer Medicine, Sir Run Run Shaw Medical Center, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
| |
Collapse
|
41
|
Zhou H, Zhang R, Men K, Tang L, Wang Y, Yang L. A Novel Systemic siDR6 Delivery System Based on DP7-C for the Treatment of Metastatic Lung Cancer. Int J Nanomedicine 2025; 20:3623-3642. [PMID: 40125426 PMCID: PMC11930241 DOI: 10.2147/ijn.s488213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 03/06/2025] [Indexed: 03/25/2025] Open
Abstract
Background The treatment of metastatic lung cancer, a common complication of many primary cancers, has historically been a significant clinical challenge. Once lung metastasis occurs, patients' survival is often significantly shortened. Therefore, prevention and treatment of lung metastases is an important aspect of cancer treatment. In this study, a simple, low-toxicity, cholesterol-modified cationic cell-penetrating peptide DP7 (DP7-C), in combination with siDR6 was used for intravenous administration for the treatment of lung metastases. Methods Initially, clinical databases were analyzed to determine the expression levels of death receptor 6 (DR6) in metastatic tumors and the correlation between DR6 expression and patient survival times. The DP7-C/siDR6 micelles were prepared by a self-assembly method. By cultivating 293T, B16F10 and LL2 cells, the in vitro experiments were performed to assess the transfection efficiency, safety and anti-cancer ability of DP7-C/siDR6, while its targeting efficiency and prevention of lungs were investigated by mouse experiments. Furthermore, the therapeutic efficacy of DP7-C/siDR6 was demonstrated in the LL2 model of lung cancer in situ, the B16F10 model of artificial lung metastasis, and the 4T1 model of spontaneous lung metastasis. Results The clinical data analysis revealed that DR6 was highly expressed in the majority of metastatic tumors and that patients with high DR6 expression exhibited significantly shorter survival times. The DP7-C/siDR6 showed high transfection efficiency, and it could inhibit tumor cell growth by suppressing the STAT3 signaling pathway. Subsequent mouse experiments demonstrated that intravenous administration of DP7-C/siDR6 resulted in efficient lung targeting. The inhibition of DR6 expression on lung endothelial cells was found to prevent metastasis-induced primary necrosis of lung endothelial cells, thereby preventing tumor metastasis. And the DP7-C/siDR6 treatment showed excellent therapeutic efficacy in the tumor models. Conclusion The systemic delivery of DP7-C micelles carrying siDR6 provide an alternative therapeutic strategy to halt cancer lung metastasis.
Collapse
Affiliation(s)
- Hongyou Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Rui Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Ke Men
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Lin Tang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Yusi Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Li Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| |
Collapse
|
42
|
Ye L, Yue WR, Shi H, Li JR, Qun YY. Case Report: Successful immune checkpoint inhibitor rechallenge after sintilimab-induced Guillain-Barré syndrome. Front Immunol 2025; 16:1546886. [PMID: 40176803 PMCID: PMC11961408 DOI: 10.3389/fimmu.2025.1546886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 02/27/2025] [Indexed: 04/04/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized hepatocellular carcinoma (HCC) treatment, while immune-related adverse events (IRAEs) pose significant challenges. We report a 60-year-old male with unresectable HCC who developed Guillain-Barré syndrome (GBS), a rare but severe neurologic complication, after three cycles of sintilimab plus bevacizumab biosimilar and conventional transarterial chemoembolization (c-TACE). The patient presented with progressive ascending weakness, reaching symmetric quadriparesis with proximal muscle strength of 2/5 in upper limbs and 1/5 in lower limbs. Following sintilimab discontinuation, treatment with intravenous immunoglobulin (2 g/kg) and oral prednisone (30 mg/day) achieved complete neurological recovery within one month. Given the patient's favorable initial tumor response and strong request, immunotherapy was cautiously reinstated using tislelizumab after thorough clinical evaluation. Following four cycles of treatment, significant tumor response enabled successful conversion surgery with major pathological response (necrosis rate >70%). With 26-month survival and no evidence of recurrence, this case demonstrates the potential feasibility of ICI rechallenge with an alternative PD-1 inhibitor following sintilimab-induced GBS. Our experience suggests that ICI-related neurological adverse events may be drug-specific rather than class-specific, potentially providing valuable treatment options for patients showing favorable tumor response despite experiencing severe IRAEs, though larger studies are needed for validation.
Collapse
Affiliation(s)
- Lin Ye
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Wan Rong Yue
- Department of Pathology, Guilin People's Hospital, Guilin, China
| | - Hao Shi
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Jian Ren Li
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Yu Ya Qun
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, China
| |
Collapse
|
43
|
Chen X, Wu X, Peng W, Liu L, Liu X, Wan X, Xu H, Zheng Y, Zhao H, Mao Y, Lu X, Sang X, Chang X, Zhou K, Pan J, Guan M, Hu D, Tan H, Zhang Y, Du S. Combined TACE with Targeted and Immunotherapy versus TACE Alone Improves DFS in HCC with MVI: A Multicenter Propensity Score Matching Study. J Hepatocell Carcinoma 2025; 12:561-577. [PMID: 40124969 PMCID: PMC11930282 DOI: 10.2147/jhc.s504016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 03/11/2025] [Indexed: 03/25/2025] Open
Abstract
Background Hepatocellular carcinoma (HCC) with microvascular invasion (MVI) is associated with high recurrence and poor survival outcomes. Although adjuvant therapies such as transcatheter arterial chemoembolization (TACE), targeted therapy, and immunotherapy show potential in improving outcomes, the optimal postoperative treatment strategy remains undetermined. This study evaluates the efficacy of different adjuvant treatments on disease-free survival (DFS) and overall survival (OS) in HCC patients with MVI following curative resection. Methods A retrospective cohort of 409 HCC patients with MVI who underwent curative resection from three clinical centers between 2017 and 2024 was analyzed. Patients were stratified into three groups: TACE alone (n=132), TACE + targeted therapy (n=58), and TACE + targeted immunotherapy (n=68). Propensity score matching (PSM) was employed to balance confounding factors. Kaplan-Meier survival curves and Cox regression models were used to assess DFS and OS. A nomogram was constructed for individualized DFS prediction. Results After PSM, both the TACE + targeted therapy and TACE + targeted immunotherapy groups exhibited significantly prolonged DFS compared to TACE alone (median DFS: 16 vs 22 and 21 months, respectively; p=0.027). No significant differences were observed in OS across the groups. The nomogram for DFS demonstrated robust predictive performance, with a C-index of 0.709 and 0.645 in the training and validation cohorts, respectively, supporting its utility in clinical decision-making. Conclusion In HCC patients with MVI, adjuvant TACE combined with targeted therapy or targeted immunotherapy significantly enhances DFS, though no OS benefit was observed. The developed nomogram provides a reliable tool for risk stratification and personalized postoperative management in this high-risk patient population.
Collapse
Affiliation(s)
- Xiaokun Chen
- Department of Liver Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100005, People’s Republic of China
- Graduate School, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100006, People’s Republic of China
| | - Xiangan Wu
- Department of Liver Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100005, People’s Republic of China
- Graduate School, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100006, People’s Republic of China
| | - Wei Peng
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, People’s Republic of China
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, People’s Republic of China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, People’s Republic of China
| | - Liguo Liu
- Second Division of Hepatopancreatobiliary Surgery, China–Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
| | - Xiao Liu
- Department of Liver Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100005, People’s Republic of China
| | - Xueshuai Wan
- Department of Liver Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100005, People’s Republic of China
| | - Haifeng Xu
- Department of Liver Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100005, People’s Republic of China
| | - Yongchang Zheng
- Department of Liver Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100005, People’s Republic of China
| | - Haitao Zhao
- Department of Liver Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100005, People’s Republic of China
| | - Yilei Mao
- Department of Liver Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100005, People’s Republic of China
| | - Xin Lu
- Department of Liver Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100005, People’s Republic of China
| | - Xinting Sang
- Department of Liver Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100005, People’s Republic of China
| | - Xiaoyan Chang
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100005, People’s Republic of China
| | - Kang Zhou
- Department of Radiology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100005, People’s Republic of China
| | - Jie Pan
- Department of Radiology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100005, People’s Republic of China
| | - Mei Guan
- Department of Medical Oncology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100005, People’s Republic of China
| | - Dandan Hu
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, People’s Republic of China
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, People’s Republic of China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, People’s Republic of China
| | - Haidong Tan
- Second Division of Hepatopancreatobiliary Surgery, China–Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
| | - Yaojun Zhang
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, People’s Republic of China
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, People’s Republic of China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, People’s Republic of China
| | - Shunda Du
- Department of Liver Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100005, People’s Republic of China
| |
Collapse
|
44
|
Hu J, Zhao S, Zuo M, Li CH, Yao W, Yang X, Xing W, Song P. Local-region treatment comparison following conversion therapy of hepatocellular carcinoma: a period and age-dependent analysis. Ther Adv Med Oncol 2025; 17:17588359251316665. [PMID: 40109509 PMCID: PMC11921006 DOI: 10.1177/17588359251316665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 01/14/2025] [Indexed: 03/22/2025] Open
Abstract
Background Transarterial chemoembolization (TACE) is a potential conversion therapeutic strategy for unresectable hepatocellular carcinoma (uHCC). However, therapeutic options following conversion therapy are still controversial. Objectives This study aimed to compare the efficacy and safety of surgical resection (SR) and microwave ablation (MWA) after TACE conversion therapy for uHCC. Design A retrospective, multi-institutional study. Methods From June 2008 to October 2022, 8842 consecutive uHCC patients underwent initial TACE at 15 hospitals were identified. Among them, 1348 eligible patients who received TACE conversion therapy were included. The propensity score matching (PSM) was applied to reduce selection bias. To explore the effect of age on conversion therapy, a therapeutic factor analysis with age change was performed. The overall survival (OS) and disease-free survival (DFS) were compared using the Kaplan-Meier method with the log-rank test. Results After PSM 1:1, 542 patients in the MWA group were matched with those in the SR group. SR demonstrated better long-term survival outcomes (median OS, 10.6 vs 5.8 years, HR:1.83, 95% CI: 1.48-2.25, p < 0.001 and median DFS, 3.2 vs 2.5 years, HR: 1.27, 95% CI:1.09-1.49, p = 0.003) than MWA. There was an improvement in the 5-year DFS rate for MWA from 17.1% during 2009-2016 to 37.3% during 2017-2022, becoming comparable to the 40.8% of SR (p = 0.129). When the uHCC patients downstage met Milan criteria, the long-term OS and DFS were comparable between two groups (both, p > 0.05). SR presents an OS advantage over MWA at the age (years) of 45-54 (p = 0.036), 55-65 (p = 0.001), and >65 (p < 0.001), except <45(p = 0.140). Conclusion MWA might be acceptable as an alternative to SR in first-line therapeutic scheme after TACE conversion therapy for uHCC, especially, for the aged <45 years cohorts.
Collapse
Affiliation(s)
- Jiahui Hu
- Institute of Medical Information, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Shu Zhao
- Department of Medical Oncology, The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Mengxuan Zuo
- Department of Minimal Invasive Intervention, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Chun Hui Li
- Department of general surgery, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China
| | - Wang Yao
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Province Guangdong, China
| | - Xinyu Yang
- Department of Radiology, The Tangshan Worker Hospital, Tangshan, HeBei, China
| | - WeiWei Xing
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Peng Song
- Department of Medical Oncology, The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Fuixing 28 Road, Beijing 100853, China
| |
Collapse
|
45
|
Shen Y, Bai X, Zhang Q, Liang X, Jin X, Zhao Z, Song W, Tan Q, Zhao R, Jia W, Gu S, Shi G, Zheng Z, Wei G, Wang Y, Fang T, Li Y, Wang Z, Yang Z, Guo S, Lin D, Wei F, Wang L, Sun X, Qin A, Xie L, Qiu Y, Bao W, Rahimian S, Singh M, Murad Y, Shang J, Chu M, Huang M, Ding J, Chen W, Ye Y, Chen Y, Li X, Liang T. Oncolytic virus VG161 in refractory hepatocellular carcinoma. Nature 2025:10.1038/s41586-025-08717-5. [PMID: 40108464 DOI: 10.1038/s41586-025-08717-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 01/30/2025] [Indexed: 03/22/2025]
Abstract
Hepatocellular carcinoma remains a life-threatening malignancy with limited therapeutic options following the failure of second-line treatments1,2. Oncolytic viruses selectively replicate in and lyse cancer cells, releasing neoantigens and stimulating systemic antitumour immunity3, offering a potential therapeutic option. Here we present the results of a multicentre phase 1 clinical trial evaluating VG161, an engineered oncolytic herpes simplex virus that expresses IL-12, IL-15, IL-15Rα and a PD-1-PD-L1-blocking fusion protein4, for safety and efficacy in patients with advanced liver cancer. VG161 was well tolerated, with no dose-limiting toxicities observed, and it demonstrated promising efficacy by reshaping the tumour immune microenvironment and re-sensitizing tumours that were previously resistant to systemic treatments. Notably, we also found that patients who had previously been sensitive to checkpoint inhibitor therapy showed enhanced efficacy with VG161 treatment. Furthermore, we developed an efficacy-prediction model based on differentially expressed genes, which successfully identified patients who were likely to benefit from VG161 and predicted prolonged overall survival. These findings position VG161 as a promising third-line therapeutic option for refractory hepatocellular carcinoma. This provides a new avenue for treatment and advances the field of oncolytic virus-based immunotherapies. ClinicalTrials.gov registration: NCT04806464 .
Collapse
Affiliation(s)
- Yinan Shen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Xueli Bai
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Qi Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Xingmei Liang
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinyan Jin
- Department of Ultrasound Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zeda Zhao
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Wei Song
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Qian Tan
- Shanghai Virogin Biotech, Shanghai, China
| | | | - William Jia
- Shanghai Virogin Biotech, Shanghai, China
- Virogin Biotech Canada, Richmond, British Columbia, Canada
- CNBG-Virogin Biotech (Shanghai), Shanghai, China
| | - Shanzhi Gu
- Department of Interventional Radiology, Hunan Cancer Hospital, Changsha, China
| | - Guoming Shi
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
| | | | - Guyue Wei
- Department of Ultrasound Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Youlei Wang
- Key Laboratory for Drug Evaluation and Clinical Research of Zhejiang Province, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tian Fang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuwei Li
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Zijun Wang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Zifan Yang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Sida Guo
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Danni Lin
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Fang Wei
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Lei Wang
- Department of Radiotherapy, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoli Sun
- Department of Radiotherapy, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Aijun Qin
- Shanghai Virogin Biotech, Shanghai, China
| | - Longshen Xie
- CNBG-Virogin Biotech (Shanghai), Shanghai, China
| | - Yeting Qiu
- Shanghai Virogin Biotech, Shanghai, China
| | | | - Shah Rahimian
- Virogin Biotech Canada, Richmond, British Columbia, Canada
| | - Manu Singh
- Virogin Biotech Canada, Richmond, British Columbia, Canada
| | - Yanal Murad
- Virogin Biotech Canada, Richmond, British Columbia, Canada
| | | | - Min Chu
- Shanghai Virogin Biotech, Shanghai, China
| | | | - Jun Ding
- Shanghai Virogin Biotech, Shanghai, China
- Virogin Biotech Canada, Richmond, British Columbia, Canada
| | - Wei Chen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yufu Ye
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yiwen Chen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Xiang Li
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China.
| |
Collapse
|
46
|
Hao MZ, Lin HL, Hu YB, Chen QZ, Chen ZX, Qiu LB, Lin DY, Zhang H, Zheng DC, Fang ZT, Liu JF. Combination therapy strategy based on selective internal radiation therapy as conversion therapy for inoperable giant hepatocellular carcinoma: A case report. World J Gastrointest Oncol 2025; 17:100861. [PMID: 40092927 PMCID: PMC11866234 DOI: 10.4251/wjgo.v17.i3.100861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 12/29/2024] [Accepted: 01/13/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) has become a growing health concern globally. Microvascular invasion and high tumor burden are key factors limiting the curative effect of selective internal radiation therapy (SIRT). CASE SUMMARY This case study reports a 49-year-old woman who was diagnosed with China Liver Cancer Staging (CNLC) IIIa HCC and > 15 cm tumor diameter. Initially, due to insufficient future liver remnant and vascular invasion, the tumor was unresectable; however, radical hepatectomy was performed after successful conversion therapy with SIRT using yttrium-90 (90Y) resin microspheres followed by hepatic arterial infusion chemotherapy (HAIC) with tyrosine kinase inhibitor (TKI) and anti-programmed death-1 (PD-1) antibody. SIRT using 90Y resin microspheres was given by the right hepatic artery and chemoembolization was simultaneously performed in the tumor's feeding vessels from the right diaphragmatic artery. HAIC was followed every three weeks with lenvatinib and tislelizumab. At 4 months post-SIRT, the tumor was downstaged to CNLC Ib and the patient successfully underwent hepatectomy. The histopathological examination of the resected specimen showed extensive necrosis. CONCLUSION This case study provides evidence for an integrated treatment strategy combining SIRT and HAIC with TKI and anti-PD-1 antibodies for patients with large HCC and microvascular invasion. Further confirmatory trials are required in the future.
Collapse
Affiliation(s)
- Ming-Zhi Hao
- Department of Tumor Interventional Radiology, Clinical Ocology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, Fujian Province, China
| | - Hai-Lan Lin
- Department of Tumor Interventional Radiology, Clinical Ocology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, Fujian Province, China
| | - Yu-Bin Hu
- Department of Tumor Interventional Radiology, Clinical Ocology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, Fujian Province, China
| | - Qi-Zhong Chen
- Department of Tumor Interventional Radiology, Clinical Ocology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, Fujian Province, China
| | - Zhang-Xian Chen
- Department of Tumor Interventional Radiology, Clinical Ocology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, Fujian Province, China
| | - Lin-Bin Qiu
- Department of Oncology and Vascular Interventional Radiology, Clinical Oncology School of Fujian Medical University, Fuzhou 350014, Fujian Province, China
| | - Duan-Yu Lin
- Department of Nuclear Medicine, Clinical Oncology School of Fujian Medical University, Fuzhou 350014, Fujian Province, China
| | - Hui Zhang
- Department of Hepatopancreatobiliary Surgical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, Fujian Province, China
| | - De-Chun Zheng
- Department of Radiology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, Fujian Province, China
| | - Zhu-Ting Fang
- Department of Oncology and Vascular Interventional Radiology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, Fujian Province, China
| | - Jing-Feng Liu
- Department of Hepatobiliary Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, Fujian Province, China
| |
Collapse
|
47
|
Chisthi MM. Current research status and future directions of hepatic arterial infusion chemotherapy for advanced hepatocellular carcinoma. World J Gastrointest Oncol 2025; 17:99068. [PMID: 40092943 PMCID: PMC11866237 DOI: 10.4251/wjgo.v17.i3.99068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/24/2024] [Accepted: 12/10/2024] [Indexed: 02/14/2025] Open
Abstract
The rapid evolution of systemic therapies for hepatocellular carcinoma (HCC), one of the most common types of liver cancer, has attracted significant attention especially to hepatic arterial infusion chemotherapy (HAIC) as a highly promising treatment approach. This method, which delivers chemotherapy directly into the liver's arterial supply, is designed to maximize the concentration of anti-cancer drugs at the tumor site while minimizing systemic side effects. Despite the potential and the encouraging results observed in various studies, HAIC has not yet achieved widespread acceptance and utilization. Sorafenib is a widely used systemic therapy that targets multiple pathways involved in tumor growth and angiogenesis, while transarterial chemoembolization (TACE) is a locoregional therapy that combines arterial embolization with chemotherapy. These treatments have been the mainstay of HCC management, yet they have limitations that HAIC may potentially overcome. This article specifically comments on the network meta-analysis that examined the current research status of HAIC, highlighting its effectiveness and safety profile in comparison to established standard treatments such as Sorafenib and TACE. Through an extensive review of existing studies, the authors conclude that patients receiving HAIC often experience better survival rates and longer periods without disease progression compared to those receiving Sorafenib or TACE.
Collapse
Affiliation(s)
- Meer M Chisthi
- Department of General Surgery, Government Medical College Pathanamthitta, Konni 689691, Kerala, India
| |
Collapse
|
48
|
Li S, Wen Q, Huang W, Qiu Z, Feng L, Yi F. A real-world study of the efficacy of second-line treatment of unresectable hepatocellular carcinoma with esophagogastric varices after progression on first-line lenvatinib combined with PD-1 inhibitor. World J Surg Oncol 2025; 23:83. [PMID: 40082982 PMCID: PMC11905572 DOI: 10.1186/s12957-025-03742-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 03/07/2025] [Indexed: 03/16/2025] Open
Abstract
PURPOSE The incidence and mortality of hepatocellular carcinoma are still high according to National Cancer Center of China. Atezolizumab plus bevacizumab has become one of the standard regimens for the first-line treatment of unresectable hepatocellular carcinoma. However, some patients still use lenvatinib in combination with immunotherapy instead of a standard "atezolizumab-bevacizumab" regimen as a lower risk of bleeding in patients with esophagogastric varices. However, there is no evidence for second-line therapy after progression on lenvatinib combined with PD-1 inhibitor in unresectable hepatocellular carcinoma till now. Herein, we aim to investigate second-line treatment among these patients. PATIENTS AND METHODS Thirty-three patients with unresectable hepatocellular carcinoma with esophagogastric varices were admitted to the Second Affiliated Hospital of Nanchang University from January 2019 to December 2023. They were treated with lenvatinib in combination with PD-1 inhibitor as first line. The efficacy was conducted according to the RECIST1.1 criteria. The endpoints included objective response rate (ORR), disease control rate (DCR), median overall survival (OS), and median progression free survival (PFS). RESULTS We identified a total of 225 patients with unresectable hepatocellular carcinoma with esophagogastric varices who received first-line lenvatinib in combination with PD-1 inhibitor, of whom 33 (14.7%) received second-line therapy. 21 patients (63.6%) were treated with regorafenib combined with PD-1 inhibitor, 6 patients (18.2%) with apatinib plus PD-1 inhibitor, 4 patients (12.1%) with bevacizumab plus PD-1 inhibitor, and the remaining 2 patients with regorafenib or sorafenib as monotherapy, respectively. Of the 33 patients, 2 (6.1%) were evaluated as partial response (PR), 16 (48.5%) had stable disease (SD), and 15 (45.4%) experienced progression (PD). The ORR was 6.1%, and the DCR was 54.6%. Median PFS was 4.5 months, median OS was 7.2 months, and the 12-month OS rate was 27.3%. Overall survival follow-up was done in 37 patients without second line treatment whose baseline levels were matched with those of the treatment group. The OS was 7.2 months in second line treatment group versus 3.0 months in control group (p = 0.04). As for different treatments in a second line, The ORR of regorafenib in combination with PD-1 inhibitor was 9.5%, the DCR was 47.6%, the median PFS was 4.2 months, and the median OS was 5.9 months. None of the patients treated with apatinib plus PD-1 inhibitor got PR, the DCR was 83.3%, the median PFS was 8.7 months, and the median OS was 9.1 months. None of the patients treated with bevacizumab plus PD-1 inhibitor got PR, the DCR was 25.0%, the median PFS was 2.2 months, and the median OS was 6.0 months. CONCLUSION The second-line treatment of unresectable hepatocellular carcinoma with esophagogastric varices after progression on first-line lenvatinib combined with PD-1 inhibitor is effective. Regorafenib or apatinib combined with PD-1 inhibitor might be the preferred options.
Collapse
Affiliation(s)
- Saifeng Li
- Department of Oncology, Second Affiliated Hospital of Nanchang University, Minde Road 1, Nanchang, 330006, P. R. China
- Jiangxi Medical College of Nanchang University, Nanchang, 330006, P. R. China
| | - Qin Wen
- Department of Oncology, Second Affiliated Hospital of Nanchang University, Minde Road 1, Nanchang, 330006, P. R. China
- Jiangxi Medical College of Nanchang University, Nanchang, 330006, P. R. China
| | - Wenwu Huang
- Department of Oncology, Second Affiliated Hospital of Nanchang University, Minde Road 1, Nanchang, 330006, P. R. China
- Jiangxi Medical College of Nanchang University, Nanchang, 330006, P. R. China
| | - Zeyu Qiu
- Department of Oncology, Second Affiliated Hospital of Nanchang University, Minde Road 1, Nanchang, 330006, P. R. China
| | - Long Feng
- Department of Oncology, Second Affiliated Hospital of Nanchang University, Minde Road 1, Nanchang, 330006, P. R. China.
| | - Fengming Yi
- Department of Oncology, Second Affiliated Hospital of Nanchang University, Minde Road 1, Nanchang, 330006, P. R. China.
| |
Collapse
|
49
|
Zhou Q, Li H, Liang Y, Li R, Wang X, Wang W, Liu M, Duan F, Huang Z. Hepatic arterial infusion chemotherapy combined with lenvatinib and immune checkpoint inhibitor versus lenvatinib for advanced hepatocellular carcinoma: a multicenter study with propensity score and coarsened exact matching. LA RADIOLOGIA MEDICA 2025:10.1007/s11547-025-01975-3. [PMID: 40072804 DOI: 10.1007/s11547-025-01975-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 02/14/2025] [Indexed: 03/14/2025]
Abstract
PURPOSE Hepatic arterial infusion chemotherapy (HAIC) combined with lenvatinib (Len) and immune checkpoint inhibitor (ICI) in treating advanced hepatocellular carcinoma (HCC) still needs further confirmation. We aimed to evaluate the efficacy of HAIC combined with Len and ICI (HAIC + Len + ICI) versus Len alone in advanced HCC. METHODS A total of 290 patients in Len group and 349 patients in HAIC + Len + ICI group were analysed. Propensity score matching (PSM), inverse probability treatment weighting (IPTW), and coarsened exact matching (CEM) analyses were used to balance the bias between two groups. Mediation analysis of treatment type in survival was performed for analysis. RESULTS The median progression-free survival (PFS) was 5.9 ± 0.2 months in Len group and 9.2 ± 0.5 months in HAIC + Len + ICI group. The HAIC + Len + ICI group demonstrated significantly better PFS than the Len group across the entire cohort (hazard ratio [HR], 0.50; 95% CI 0.43-0.60; P < 0.001). This advantage in PFS was sustained in the PSM, IPTW, and CEM cohorts. HAIC + Len + ICI group also showed better overall survival (OS) than the Len group (HR, 0.38; 95% CI 0.31-0.46; P < 0.001). The OS was also superior in the PSM, IPTW, and CEM cohorts. The objective response rate (ORR) in HAIC + Len + ICI group was twice as high as that in Len group. Further mediation analysis showed tumor response at 3 and 6 months had different mediation effect on survival. CONCLUSIONS HAIC combined with Len and ICI showed improved better OS and PFS than Len alone. This triple therapy could be considered as a first-line treatment for advanced HCC.
Collapse
Affiliation(s)
- Qunfang Zhou
- Department of Interventional Radiology, 5th Medical Center of Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Hui Li
- Department of Minimally Invasive Interventional Radiology, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Dongfeng East Road 651, Guangzhou, 510260, Guangdong Province, China
| | - Ye Liang
- Department of Interventional Radiology, 5th Medical Center of Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Ruixia Li
- Department of Liver Surgery, The First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, China
| | - Xiaohui Wang
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, 61 Jiefang West Road, Changsha, Hunan Province, China
| | - Wei Wang
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Renmin Road No. 2, Jinzhou, 121000, Liaoning Province, China
| | - Mingyu Liu
- Department of Interventional Radiology, The Affiliated Shunde Hospital of Jinan University, Guizhou East Road 50, Foshan, Guangdong Province, China
| | - Feng Duan
- Department of Interventional Radiology, 5th Medical Center of Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China.
| | - Zhimei Huang
- Department of Minimally Invasive Interventional Radiology, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Dongfeng East Road 651, Guangzhou, 510260, Guangdong Province, China.
| |
Collapse
|
50
|
Liu F, Xiao L, Zhao L, Tao Y, Huang D, Chen Z, He C, Wu C. Prostate-specific membrane antigen-targeting radiopharmaceuticals: a new frontier in hepatic malignancies. Front Oncol 2025; 15:1547459. [PMID: 40123907 PMCID: PMC11926431 DOI: 10.3389/fonc.2025.1547459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 02/18/2025] [Indexed: 03/25/2025] Open
Abstract
Background/Objectives Prostate-specific membrane antigen (PSMA) is overexpressed in prostate hypercellularity, making it an effective target for molecular imaging and therapy of prostate cancer. PSMA is expressed in the neovasculature of hepatic malignancies and regulates tumor cell invasion and angiogenesis. The diagnosis and treatment of hepatic malignancies remain challenging. Thus, radiopharmaceuticals targeting PSMA are gaining prominence in the treatment of hepatic malignancies. Therefore, this review aims to discuss the applications of PSMA-targeting radiopharmaceuticals in hepatic malignant tumors, focusing on hepatocellular carcinoma (HCC), to assess their value as a diagnostic and therapeutic agent for hepatic malignancies. Methods The potentials of PSMA-targeting radiopharmaceuticals for diagnostic and therapeutic use in hepatic malignancies were investigated. Moreover, their characteristics, diagnostic and therapeutic efficacies, and potential synergies when used in conjunction with other therapeutic modalities were elucidated. Results Computed tomography (CT) and magnetic resonance imaging (MRI) are the most common imaging modalities in clinical practice; however, their sensitivity is not optimal. PSMA positron emission tomography/CT can be used as a complementary modality to conventional imaging for characterizing lesions, staging and/or re-staging HCC, and assessing treatment response when conventional imaging results are unclear. Moreover, most patients with HCC are diagnosed at an advanced stage in which treatment options are limited. Hence, PSMA-based radioligand therapy serves as a promising alternative treatment when multiple treatments fail. Conclusions Further research and clinical transformation are required to effectively diagnose and treat HCC via PSMA targeting. This will have significant clinical application prospects in primary and secondary hepatic malignancies.
Collapse
Affiliation(s)
- Fucen Liu
- National Health Commission (NHC) Key Laboratory of Nuclear Technology Medical Transformation (MIANYANG CENTRAL HOSPITAL), Mianyang, China
- Department of Nuclear Medicine, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Liming Xiao
- National Health Commission (NHC) Key Laboratory of Nuclear Technology Medical Transformation (MIANYANG CENTRAL HOSPITAL), Mianyang, China
- Department of Nuclear Medicine, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Ling Zhao
- National Health Commission (NHC) Key Laboratory of Nuclear Technology Medical Transformation (MIANYANG CENTRAL HOSPITAL), Mianyang, China
- Department of Nuclear Medicine, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
- Institute of Basic Medicine, North Sichuan Medical College, Nanchong, China
| | - Yi Tao
- National Health Commission (NHC) Key Laboratory of Nuclear Technology Medical Transformation (MIANYANG CENTRAL HOSPITAL), Mianyang, China
- Department of Nuclear Medicine, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Dan Huang
- National Health Commission (NHC) Key Laboratory of Nuclear Technology Medical Transformation (MIANYANG CENTRAL HOSPITAL), Mianyang, China
- Department of Nuclear Medicine, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Zhengguo Chen
- National Health Commission (NHC) Key Laboratory of Nuclear Technology Medical Transformation (MIANYANG CENTRAL HOSPITAL), Mianyang, China
- Department of Nuclear Medicine, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Chuandong He
- National Health Commission (NHC) Key Laboratory of Nuclear Technology Medical Transformation (MIANYANG CENTRAL HOSPITAL), Mianyang, China
- Department of Nuclear Medicine, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Chunyan Wu
- National Health Commission (NHC) Key Laboratory of Nuclear Technology Medical Transformation (MIANYANG CENTRAL HOSPITAL), Mianyang, China
- Department of Nuclear Medicine, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| |
Collapse
|